1
|
Vazquez Cegla AJ, Jones KT, Cui G, Cottrill KA, Koval M, McCarty NA. Effects of hyperglycemia on airway epithelial barrier function in WT and CF 16HBE cells. Sci Rep 2024; 14:25095. [PMID: 39443580 PMCID: PMC11500396 DOI: 10.1038/s41598-024-76526-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Cystic fibrosis related diabetes (CFRD), the main co-morbidity in cystic fibrosis (CF), is associated with higher rates of lung function decline. We hypothesize that airway epithelial barrier function is impaired in CF and is further exacerbated under hyperglycemia, worsening pulmonary outcomes. Using 16HBE cells, we studied the effects of hyperglycemia in airway epithelial barrier function. Results show increased paracellular dye flux in CF cells in response to insulin under hyperglycemia. Gene expression experiments identified claudin-4 (CLDN4) as a key tight junction protein dysregulated in CF cells. CLDN4 protein localization by confocal microscopy showed that CLDN4 was tightly localized at tight junctions in WT cells, which did not change under hyperglycemia. ln contrast, CLDN4 was less well-localized in CF cells at normal glucose and localization was worsened under hyperglycemia. Treatment with highly effective modulator compounds (ETI) reversed this trend, and CFTR rescue was not affected by insulin or hyperglycemia. Bulk RNA sequencing showed differences in transcriptional responses in CF compared to WT cells under normal or high glucose, highlighting promising targets for future investigation. One of these targets is protein tyrosine phosphatase receptor type G (PTPRG), which has been previously found to play a role in defective Akt signaling and insulin resistance.
Collapse
Affiliation(s)
- Analia J Vazquez Cegla
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
- Emory+Children's Cystic Fibrosis Center of Excellence, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Kymry T Jones
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
- Emory+Children's Cystic Fibrosis Center of Excellence, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Guiying Cui
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Kirsten A Cottrill
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
- Emory+Children's Cystic Fibrosis Center of Excellence, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Michael Koval
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nael A McCarty
- Division of Pulmonology, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Emory+Children's Cystic Fibrosis Center of Excellence, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
2
|
Karetnikova ES, Livanova AA, Fedorova AA, Markov AG. Early Radiation-Induced Changes in Lung Tissue and Intercellular Junctions: Implications for Tissue Repair and Fibrosis. PATHOPHYSIOLOGY 2024; 31:531-544. [PMID: 39449521 PMCID: PMC11503413 DOI: 10.3390/pathophysiology31040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/13/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024] Open
Abstract
Early changes in lung tissue following ionizing radiation (IR) initiate processes that may lead to either regeneration or fibrosis. Intercellular junction proteins play a crucial role in the organization and function of epithelial tissues, both under normal conditions and after injuries. Alterations in the expression and localization of these proteins can influence the fate of epithelial cells. This study aims to investigate the effects of IR on lung tissue structure, as well as on the levels and distribution of intercellular junction proteins. Wistar rats were subjected to total X-ray irradiation at doses of 2 and 10 Gy. Lung tissue samples were collected for Western blot and histological analysis 72 h post-IR. IR at doses of 2 and 10 Gy led to structural changes in lung tissue and elevated levels of E-cadherin. The 10 Gy IR resulted in increased claudin-4 and occludin in lung parenchyma, decreased claudin-8 and claudin-12 in bronchial epithelium and endothelium, and suppression of apoptosis. Data evaluation indicated that alterations in the protein composition of intercellular junctions are essential processes in lung tissue at early stages after IR, and at least some of these alterations are associated with adaptation.
Collapse
Affiliation(s)
- Ekaterina S. Karetnikova
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
- Interoception Laboratory, Pavlov Institute of Physiology RAS, 199034 St. Petersburg, Russia
| | - Alexandra A. Livanova
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Arina A. Fedorova
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Alexander G. Markov
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
- Interoception Laboratory, Pavlov Institute of Physiology RAS, 199034 St. Petersburg, Russia
| |
Collapse
|
3
|
Erramilli SK, Dominik PK, Ogbu CP, Kossiakoff AA, Vecchio AJ. Structural and biophysical insights into targeting of claudin-4 by a synthetic antibody fragment. Commun Biol 2024; 7:733. [PMID: 38886509 PMCID: PMC11183071 DOI: 10.1038/s42003-024-06437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Claudins are a 27-member family of ~25 kDa membrane proteins that integrate into tight junctions to form molecular barriers at the paracellular spaces between endothelial and epithelial cells. As the backbone of tight junction structure and function, claudins are attractive targets for modulating tissue permeability to deliver drugs or treat disease. However, structures of claudins are limited due to their small sizes and physicochemical properties-these traits also make therapy development a challenge. Here we report the development of a synthetic antibody fragment (sFab) that binds human claudin-4 and the determination of a high-resolution structure of it bound to claudin-4/enterotoxin complexes using cryogenic electron microscopy. Structural and biophysical results reveal this sFabs mechanism of select binding to human claudin-4 over other homologous claudins and establish the ability of sFabs to bind hard-to-target claudins to probe tight junction structure and function. The findings provide a framework for tight junction modulation by sFabs for tissue-selective therapies.
Collapse
Affiliation(s)
- Satchal K Erramilli
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Pawel K Dominik
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
- Pfizer, San Diego, CA, 92121, USA
| | - Chinemerem P Ogbu
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Department of Structural Biology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Alex J Vecchio
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
- Department of Structural Biology, University at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
4
|
Sun W, Wu W, Fang X, Ge X, Zhang Y, Han J, Guo X, Zhou L, Yang H. Disruption of pulmonary microvascular endothelial barrier by dysregulated claudin-8 and claudin-4: uncovered mechanisms in porcine reproductive and respiratory syndrome virus infection. Cell Mol Life Sci 2024; 81:240. [PMID: 38806818 PMCID: PMC11133251 DOI: 10.1007/s00018-024-05282-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
The pulmonary endothelium is a dynamic and metabolically active monolayer of endothelial cells. Dysfunction of the pulmonary endothelial barrier plays a crucial role in the acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), frequently observed in the context of viral pneumonia. Dysregulation of tight junction proteins can lead to the disruption of the endothelial barrier and subsequent leakage. Here, the highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) served as an ideal model for studying ALI and ARDS. The alveolar lavage fluid of pigs infected with HP-PRRSV, and the supernatant of HP-PRRSV infected pulmonary alveolar macrophages were respectively collected to treat the pulmonary microvascular endothelial cells (PMVECs) in Transwell culture system to explore the mechanism of pulmonary microvascular endothelial barrier leakage caused by viral infection. Cytokine screening, addition and blocking experiments revealed that proinflammatory cytokines IL-1β and TNF-α, secreted by HP-PRRSV-infected macrophages, disrupt the pulmonary microvascular endothelial barrier by downregulating claudin-8 and upregulating claudin-4 synergistically. Additionally, three transcription factors interleukin enhancer binding factor 2 (ILF2), general transcription factor III C subunit 2 (GTF3C2), and thyroid hormone receptor-associated protein 3 (THRAP3), were identified to accumulate in the nucleus of PMVECs, regulating the transcription of claudin-8 and claudin-4. Meanwhile, the upregulation of ssc-miR-185 was found to suppress claudin-8 expression via post-transcriptional inhibition. This study not only reveals the molecular mechanisms by which HP-PRRSV infection causes endothelial barrier leakage in acute lung injury, but also provides novel insights into the function and regulation of tight junctions in vascular homeostasis.
Collapse
Affiliation(s)
- Weifeng Sun
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- China Institute of Veterinary Drug Control, Beijing, 100081, People's Republic of China
| | - Weixin Wu
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Xinyu Fang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Xinna Ge
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Yongning Zhang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Jun Han
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Xin Guo
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Lei Zhou
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
| | - Hanchun Yang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
| |
Collapse
|
5
|
Lindstedt S, Wang Q, Niroomand A, Stenlo M, Hyllen S, Pierre L, Olm F, Bechet NB. High resolution fluorescence imaging of the alveolar scaffold as a novel tool to assess lung injury. Sci Rep 2024; 14:6662. [PMID: 38509285 PMCID: PMC10954697 DOI: 10.1038/s41598-024-57313-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/17/2024] [Indexed: 03/22/2024] Open
Abstract
Acute lung injury (ALI) represents an aetiologically diverse form of pulmonary damage. Part of the assessment and diagnosis of ALI depends on skilled observer-based scoring of brightfield microscopy tissue sections. Although this readout is sufficient to determine gross alterations in tissue structure, its categorical scores lack the sensitivity to describe more subtle changes in lung morphology. To generate a more sensitive readout of alveolar perturbation we carried out high resolution immunofluorescence imaging on 200 μm lung vibratome sections from baseline and acutely injured porcine lung tissue, stained with a tomato lectin, Lycopersicon Esculentum Dylight-488. With the ability to resolve individual alveoli along with their inner and outer wall we generated continuous readouts of alveolar wall thickness and circularity. From 212 alveoli traced from 10 baseline lung samples we established normal distributions for alveolar wall thickness (27.37; 95% CI [26.48:28.26]) and circularity (0.8609; 95% CI [0.8482:0.8667]) in healthy tissue. Compared to acutely injured lung tissue baseline tissue exhibited a significantly lower wall thickness (26.86 ± 0.4998 vs 50.55 ± 4.468; p = 0.0003) and higher degree of circularityϕ≤ (0.8783 ± 0.01965 vs 0.4133 ± 0.04366; p < 0.0001). These two components were subsequently combined into a single more sensitive variable, termed the morphological quotient (MQ), which exhibited a significant negative correlation (R2 = 0.9919, p < 0.0001) with the gold standard of observer-based scoring. Through the utilisation of advanced light imaging we show it is possible to generate sensitive continuous datasets describing fundamental morphological changes that arise in acute lung injury. These data represent valuable new analytical tools that can be used to precisely benchmark changes in alveolar morphology both in disease/injury as well as in response to treatment/therapy.
Collapse
Affiliation(s)
- Sandra Lindstedt
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
- Department of Clinical Sciences, Lund University, Lund, Sweden.
- Lund Stem Cell Centre, Lund University, Lund, Sweden.
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden.
| | - Qi Wang
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Anna Niroomand
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Centre, Lund University, Lund, Sweden
| | - Martin Stenlo
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Centre, Lund University, Lund, Sweden
- Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Snejana Hyllen
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Centre, Lund University, Lund, Sweden
- Department of Cardiothoracic Anaesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Leif Pierre
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Centre, Lund University, Lund, Sweden
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Franziska Olm
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Centre, Lund University, Lund, Sweden
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - Nicholas B Bechet
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
- Department of Clinical Sciences, Lund University, Lund, Sweden.
- Lund Stem Cell Centre, Lund University, Lund, Sweden.
| |
Collapse
|
6
|
Erramilli SK, Dominik PK, Ogbu CP, Kossiakoff AA, Vecchio AJ. Cryo-EM structures of a synthetic antibody against 22 kDa claudin-4 reveal its complex with Clostridium perfringens enterotoxin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544689. [PMID: 37398044 PMCID: PMC10312657 DOI: 10.1101/2023.06.12.544689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Claudins are a family of ∼25 kDa membrane proteins that integrate into tight junctions to form molecular barriers at the paracellular spaces between endothelial and epithelial cells. Humans have 27 subtypes, which homo- and hetero-oligomerize to impart distinct properties and physiological functions to tissues and organs. As the structural and functional backbone of tight junctions, claudins are attractive targets for therapeutics capable of modulating tissue permeability to deliver drugs or treat disease. However, structures of claudins are limited due to their small sizes and physicochemical properties-these traits also make therapy development a challenge. We have developed a synthetic antibody fragment (sFab) that binds human claudin-4 and used it to resolve structures of its complex with Clostridium perfringens enterotoxin (CpE) using cryogenic electron microscopy (cryo-EM). The resolution of the structures reveals the architectures of 22 kDa claudin-4, the 14 kDa C-terminal domain of CpE, and the mechanism by which this sFab binds claudins. Further, we elucidate the biochemical and biophysical bases of sFab binding and demonstrate that this molecule exhibits subtype-selectivity by assaying homologous claudins. Our results provide a framework for developing sFabs against hard-to-target claudins and establishes the utility of sFabs as fiducial markers for determining cryo-EM structures of this small membrane protein family at resolutions that surpass X-ray crystallography. Taken together, this work highlights the ability of sFabs to elucidate claudin structure and function and posits their potential as therapeutics for modulating tight junctions by targeting specific claudin subtypes.
Collapse
|
7
|
He T, Jin Z, Hu W, Xia X, Li D, Yao W, Li G, Zhou X, Song G. Tetrahydrocurcumin (THC) enhanced the clearance of Cryptococcus deneoformans during infection in vivo. Antonie Van Leeuwenhoek 2023; 116:565-576. [PMID: 37186068 DOI: 10.1007/s10482-023-01830-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023]
Abstract
Cryptococcal species often cause lung infections and are the main cause of fungal meningitis. Claudin-4 appears to be a major structural component that maintains a tight alveolar barrier and prevents fluid and electrolyte leakage into the alveolar space. We aimed to determine whether S7-tetrahydrocurcumin (THC) could clearance of C. deneoformans and regulate claudin-4 expression during C. deneoformans infection. We investigated the effect of THC on C. deneoformans infection and its possible mechanism in vivo. Transmission electron microscopy was used to observe the ultrastructure of the lung tissue and the invasion of Cryptococcus. To clarify the effect of THC, we examined claudin-4, c-Jun, and Smad2 expression. We also measured claudin-4 expression in pulmonary specimens from clinical patients. THC reduced cryptococcal cell invasion in the lungs, improved alveolar exudation, and reduced inflammation. Pretreatment with THC suppressed c-Jun and Smad2 expression, resulting in significantly increased claudin-4 levels. In contrast, the expression of claudin-4 in clinical specimens from patients with cryptococcal infection was higher than that in normal specimens. THC enhanced the clearance of C. deneoformans during infection in vivo. We investigated the expression of claudin-4 and the possible mechanism of THC against C. deneoformans infection.
Collapse
Affiliation(s)
- Tianli He
- Department of Radiotherapy, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou, 313100, Zhejiang, China
| | - Zhiran Jin
- Department of Surgery, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou, 313100, Zhejiang, China
| | - Wei Hu
- Department of Radiotherapy, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou, 313100, Zhejiang, China
| | - Xiaoxue Xia
- Department of Infectious Diseases Department of Respiratory, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou , 313100, Zhejiang, China
| | - Donghui Li
- Department of Neurology, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou, 313100, Zhejiang, China
| | - Weiyun Yao
- Department of Surgery, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou, 313100, Zhejiang, China
| | - Guangnan Li
- Department of Respiratory Medicine, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, 313100, Zhejiang, China
- Department of Respiratory Medicine, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou, 313100, Zhejiang, China
| | - Xuefeng Zhou
- Department of Respiratory Medicine, Changxing People's Hospital, No. 66, Taihu Road, Changxing, Huzhou, 313100, Zhejiang, China
| | - Guoqiang Song
- Department of Respiratory Medicine, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, 313100, Zhejiang, China.
| |
Collapse
|
8
|
Liu Y, Xu J, Shi J, Zhang Y, Ma Y, Zhang Q, Su Z, Zhang Y, Hong S, Hu G, Chen Z, Jia G. Effects of short-term high-concentration exposure to PM 2.5 on pulmonary tissue damage and repair ability as well as innate immune events. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 319:121055. [PMID: 36632972 DOI: 10.1016/j.envpol.2023.121055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/15/2022] [Accepted: 01/08/2023] [Indexed: 06/17/2023]
Abstract
Short-term heavy air pollution still occurs frequently worldwide, especially during the winter heating period in some developing countries, which is usually accompanied by the temporary explosive growth of PM2.5. The pulmonary damage caused by PM2.5 exposure has been determined, but there have been few studies on the repair ability after the cessation of exposure and the important role of innate immune events. This study established a short-term (30 days) high-concentration (15 mg/kg body weight) PM2.5 exposure and recovery (15 days of exposure cessation) model by intratracheal instillation. The results showed that short-term PM2.5 exposure increased the content of collagen fiber in rat lung tissue, which was significantly repaired after recovery by 15 days of exposure cessation. Meanwhile, exposure to PM2.5 also caused changes in lung epithelial function, macrophage polarization and cell autophagy function. Most of these changes could be restored or reversed to a certain extent after recovery. However, there were also some biomarkers, including CLDN18.1, SP-A, SP-D, iNOS, CD206, Beclin1, p62 and LC3B, that were still significantly different between the exposure and control groups after recovery, suggesting that some toxic effects, especially epithelial function damage, were not completely repaired. In addition, there was a significant correlation between pulmonary fibrosis and innate immunity. The present study demonstrated that short-term high-concentration exposure to PM2.5 could cause temporary lung tissue damage and related innate immune events in rats, and the repair ability existed after the cessation of exposure, but part of the damage that required special attention still persisted.
Collapse
Affiliation(s)
- Yu Liu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Jiayu Xu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Jiaqi Shi
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Yi Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Ying Ma
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Qiaojian Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Zekang Su
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Yali Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Shiyi Hong
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| | - Guiping Hu
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, China
| | - Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China.
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing, 100083, China
| |
Collapse
|
9
|
Godbole NM, Chowdhury AA, Chataut N, Awasthi S. Tight Junctions, the Epithelial Barrier, and Toll-like Receptor-4 During Lung Injury. Inflammation 2022; 45:2142-2162. [PMID: 35779195 PMCID: PMC9649847 DOI: 10.1007/s10753-022-01708-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/31/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022]
Abstract
Lung epithelium is constantly exposed to the environment and is critically important for the orchestration of initial responses to infectious organisms, toxins, and allergic stimuli, and maintenance of normal gaseous exchange and pulmonary function. The integrity of lung epithelium, fluid balance, and transport of molecules is dictated by the tight junctions (TJs). The TJs are formed between adjacent cells. We have focused on the topic of the TJ structure and function in lung epithelial cells. This review includes a summary of the last twenty years of literature reports published on the disrupted TJs and epithelial barrier in various lung conditions and expression and regulation of specific TJ proteins against pathogenic stimuli. We discuss the molecular signaling and crosstalk among signaling pathways that control the TJ structure and function. The Toll-like receptor-4 (TLR4) recognizes the pathogen- and damage-associated molecular patterns released during lung injury and inflammation and coordinates cellular responses. The molecular aspects of TLR4 signaling in the context of TJs or the epithelial barrier are not fully known. We describe the current knowledge and possible networking of the TLR4-signaling with cellular and molecular mechanisms of TJs, lung epithelial barrier function, and resistance to treatment strategies.
Collapse
Affiliation(s)
- Nachiket M Godbole
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Asif Alam Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Neha Chataut
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Shanjana Awasthi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
10
|
Zheng Y, Zheng M, Shao J, Jiang C, Shen J, Tao R, Deng Y, Xu Y, Lu Y. Upregulation of claudin‑4 by Chinese traditional medicine Shenfu attenuates lung tissue damage by acute lung injury aggravated by acute gastrointestinal injury. PHARMACEUTICAL BIOLOGY 2022; 60:1981-1993. [PMID: 36226770 PMCID: PMC9578493 DOI: 10.1080/13880209.2022.2128824] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 08/16/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
CONTEXT Many studies have explored new methods to cure acute lung injury (ALI); however, none of those methods could significantly change the high mortality rate of ALI. Shenfu is a Chinese traditional medicine that might be effective against ALI. OBJECTIVE Our study explores the therapeutic potential of Shenfu in ALI. MATERIALS AND METHODS Male C57BL/6 mice were assigned to control, lipopolysaccharide (LPS) (500 µg/100 μL per mouse), and LPS + Shenfu (30 mL/kg) groups. Shenfu (10 µL/mL) was added to LPS (10 µg/mL) treated MLE-12 cells for 48 h in vitro. Male C57BL/6 mice were divided into four groups: LPS, LPS + 3% dextran sulphate sodium (DSS), 3% DSS + Shenfu, and LPS + 3% DSS + Shenfu. RESULTS Compared with the ALI group, Shenfu reduced wet/dry weight ratio (19.8%, 36.2%), and reduced the IL-2 (40.9%, 61.6%), IFN-γ (43.5%, 53.3%) TNF-α (54.1%, 42.1%), IL-6 (54.8%,70%), and IL-1β (39.9%, 65.1%), reduced serum uric acid (18.8%, 48.7%) and creatinine (17.4%, 41.1%). Moreover, Shenfu enhanced cell viability (17.2%, 59.9%) and inhibited cell apoptosis (63.0%) and p38/ERK phosphorylation in in vitro cultured epithelial cells with LPS stimulation. Mechanistically, Shenfu mediated the protective effect by upregulating claudin-4 expression. In addition, Shenfu could protect against both lung and intestinal epithelial damage in acute gastrointestinal injury-exacerbated ALI. DISCUSSION AND CONCLUSIONS Taken together, the results revealed the therapeutic effect and the underlying mechanism of Shenfu injection in an ALI in mouse model, indicating its clinical potential to treat patients with ALI.
Collapse
Affiliation(s)
- Yueliang Zheng
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- Emergency & Intensive Care Unit Center, Department of Emergency Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Mian Zheng
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Shao
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Chengxing Jiang
- Emergency & Intensive Care Unit Center, Department of Emergency Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jian Shen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Rujia Tao
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yuqin Deng
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yingge Xu
- Emergency & Intensive Care Unit Center, Department of Emergency Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yuanqiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
11
|
Gao N, Rezaee F. Airway Epithelial Cell Junctions as Targets for Pathogens and Antimicrobial Therapy. Pharmaceutics 2022; 14:2619. [PMID: 36559113 PMCID: PMC9786141 DOI: 10.3390/pharmaceutics14122619] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Intercellular contacts between epithelial cells are established and maintained by the apical junctional complexes (AJCs). AJCs conserve cell polarity and build epithelial barriers to pathogens, inhaled allergens, and environmental particles in the respiratory tract. AJCs consist of tight junctions (TJs) and adherens junctions (AJs), which play a key role in maintaining the integrity of the airway barrier. Emerging evidence has shown that different microorganisms cause airway barrier dysfunction by targeting TJ and AJ proteins. This review discusses the pathophysiologic mechanisms by which several microorganisms (bacteria and viruses) lead to the disruption of AJCs in airway epithelial cells. We present recent progress in understanding signaling pathways involved in the formation and regulation of cell junctions. We also summarize the potential chemical inhibitors and pharmacological approaches to restore the integrity of the airway epithelial barrier. Understanding the AJCs-pathogen interactions and mechanisms by which microorganisms target the AJC and impair barrier function may further help design therapeutic innovations to treat these infections.
Collapse
Affiliation(s)
- Nannan Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Fariba Rezaee
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children’s, Cleveland, OH 44195, USA
| |
Collapse
|
12
|
Felix NS, Maia LA, Rocha NN, Rodrigues GC, Medeiros M, da Silva LA, Baldavira CM, Fernezlian SDM, Eher EM, Capelozzi VL, Malbrain MLNG, Pelosi P, Rocco PRM, Silva PL. Biological impact of restrictive and liberal fluid strategies at low and high PEEP levels on lung and distal organs in experimental acute respiratory distress syndrome. Front Physiol 2022; 13:992401. [PMID: 36388107 PMCID: PMC9663484 DOI: 10.3389/fphys.2022.992401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Fluid regimens in acute respiratory distress syndrome (ARDS) are conflicting. The amount of fluid and positive end-expiratory pressure (PEEP) level may interact leading to ventilator-induced lung injury (VILI). We therefore evaluated restrictive and liberal fluid strategies associated with low and high PEEP levels with regard to lung and kidney damage, as well as cardiorespiratory function in endotoxin-induced ARDS. Methods: Thirty male Wistar rats received an intratracheal instillation of Escherichia coli lipopolysaccharide. After 24 h, the animals were anesthetized, protectively ventilated (VT = 6 ml/kg), and randomized to restrictive (5 ml/kg/h) or liberal (40 ml/kg/h) fluid strategies (Ringer lactate). Both groups were then ventilated with PEEP = 3 cmH2O (PEEP3) and PEEP = 9 cmH2O (PEEP9) for 1 h (n = 6/group). Echocardiography, arterial blood gases, and lung mechanics were evaluated throughout the experiments. Histologic analyses were done on the lungs, and molecular biology was assessed in lungs and kidneys using six non-ventilated animals with no fluid therapy. Results: In lungs, the liberal group showed increased transpulmonary plateau pressure compared with the restrictive group (liberal, 23.5 ± 2.9 cmH2O; restrictive, 18.8 ± 2.3 cmH2O, p = 0.046) under PEEP = 9 cmH2O. Gene expression associated with inflammation (interleukin [IL]-6) was higher in the liberal-PEEP9 group than the liberal-PEEP3 group (p = 0.006) and restrictive-PEEP9 (p = 0.012), Regardless of the fluid strategy, lung mechanical power and the heterogeneity index were higher, whereas birefringence for claudin-4 and zonula-ocludens-1 gene expression were lower in the PEEP9 groups. Perivascular edema was higher in liberal groups, regardless of PEEP levels. Markers related to damage to epithelial cells [club cell secreted protein (CC16)] and the extracellular matrix (syndecan) were higher in the liberal-PEEP9 group than the liberal-PEEP3 group (p = 0.010 and p = 0.024, respectively). In kidneys, the expression of IL-6 and neutrophil gelatinase-associated lipocalin was higher in PEEP9 groups, regardless of the fluid strategy. For the liberal strategy, PEEP = 9 cmH2O compared with PEEP = 3 cmH2O reduced the right ventricle systolic volume (37%) and inferior vena cava collapsibility index (45%). Conclusion: The combination of a liberal fluid strategy and high PEEP led to more lung damage. The application of high PEEP, regardless of the fluid strategy, may also be deleterious to kidneys.
Collapse
Affiliation(s)
- Nathane S. Felix
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ligia A. Maia
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nazareth N. Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Gisele C. Rodrigues
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mayck Medeiros
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia A. da Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila M. Baldavira
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Esmeralda M. Eher
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vera L. Capelozzi
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Manu L. N. G. Malbrain
- First Department of Anesthesiology and Intensive Therapy, Medical University of Lublin, Lublin, Poland
| | - Paolo Pelosi
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
- San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L. Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Pedro L. Silva,
| |
Collapse
|
13
|
Liu J, Dean DA. Gene Therapy for Acute Respiratory Distress Syndrome. Front Physiol 2022; 12:786255. [PMID: 35111077 PMCID: PMC8801611 DOI: 10.3389/fphys.2021.786255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating clinical syndrome that leads to acute respiratory failure and accounts for over 70,000 deaths per year in the United States alone, even prior to the COVID-19 pandemic. While its molecular details have been teased apart and its pathophysiology largely established over the past 30 years, relatively few pharmacological advances in treatment have been made based on this knowledge. Indeed, mortality remains very close to what it was 30 years ago. As an alternative to traditional pharmacological approaches, gene therapy offers a highly controlled and targeted strategy to treat the disease at the molecular level. Although there is no single gene or combination of genes responsible for ARDS, there are a number of genes that can be targeted for upregulation or downregulation that could alleviate many of the symptoms and address the underlying mechanisms of this syndrome. This review will focus on the pathophysiology of ARDS and how gene therapy has been used for prevention and treatment. Strategies for gene delivery to the lung, such as barriers encountered during gene transfer, specific classes of genes that have been targeted, and the outcomes of these approaches on ARDS pathogenesis and resolution will be discussed.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| | - David A. Dean
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
14
|
Liu J, Dean DA. Gene transfer of MRCKα rescues lipopolysaccharide-induced acute lung injury by restoring alveolar capillary barrier function. Sci Rep 2021; 11:20862. [PMID: 34675326 PMCID: PMC8531330 DOI: 10.1038/s41598-021-99897-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/27/2021] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury/Acute Respiratory Distress Syndrome (ALI/ARDS) is characterized by alveolar edema accumulation with reduced alveolar fluid clearance (AFC), alveolar-capillary barrier disruption, and substantial inflammation, all leading to acute respiratory failure. Enhancing AFC has long been considered one of the primary therapeutic goals in gene therapy treatments for ARDS. We previously showed that electroporation-mediated gene delivery of the Na+, K+-ATPase β1 subunit not only increased AFC, but also restored alveolar barrier function through upregulation of tight junction proteins, leading to treatment of LPS-induced ALI in mice. We identified MRCKα as an interaction partner of β1 which mediates this upregulation in cultured alveolar epithelial cells. In this study, we investigate whether electroporation-mediated gene transfer of MRCKα to the lungs can attenuate LPS-induced acute lung injury in vivo. Compared to mice that received a non-expressing plasmid, those receiving the MRCKα plasmid showed attenuated LPS-increased pulmonary edema and lung leakage, restored tight junction protein expression, and improved overall outcomes. Interestingly, gene transfer of MRCKα did not alter AFC rates. Studies using both cultured microvascular endothelial cells and mice suggest that β1 and MRCKα upregulate junctional complexes in both alveolar epithelial and capillary endothelial cells, and that one or both barriers may be positively affected by our approach. Our data support a model of treatment for ALI/ARDS in which improvement of alveolar-capillary barrier function alone may be of more benefit than improvement of alveolar fluid clearance.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA.,Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - David A Dean
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA. .,Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA.
| |
Collapse
|
15
|
Smyth T, Georas SN. Effects of ozone and particulate matter on airway epithelial barrier structure and function: a review of in vitro and in vivo studies. Inhal Toxicol 2021; 33:177-192. [PMID: 34346824 DOI: 10.1080/08958378.2021.1956021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The airway epithelium represents a crucial line of defense against the spread of inhaled pathogens. As the epithelium is the first part of the body to be exposed to the inhaled environment, it must act as both a barrier to and sentinel against any inhaled agents. Despite its vital role in limiting the spread of inhaled pathogens, the airway epithelium is also regularly exposed to air pollutants which disrupt its normal function. Here we review the current understanding of the structure and composition of the airway epithelial barrier, as well as the impact of inhaled pollutants, including the reactive gas ozone and particulate matter, on epithelial function. We discuss the current in vitro, rodent model, and human exposure findings surrounding the impact of various inhaled pollutants on epithelial barrier function, mucus production, and mucociliary clearance. Detailed information on how inhaled pollutants impact epithelial structure and function will further our understanding of the adverse health effects of air pollution exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steve N Georas
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
16
|
Zhang Y, Zhang L, Chen W, Zhang Y, Wang X, Dong Y, Zhang W, Lin X. Shp2 regulates PM2.5-induced airway epithelial barrier dysfunction by modulating ERK1/2 signaling pathway. Toxicol Lett 2021; 350:62-70. [PMID: 34252507 DOI: 10.1016/j.toxlet.2021.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 11/27/2022]
Abstract
The impact of fine particulate matter (PM2.5) on public health has received increasing attention. Through various biochemical mechanisms, PM2.5 alters the normal structure and function of the airway epithelium, causing epithelial barrier dysfunction. Src homology domain 2-containing protein tyrosine phosphatase 2 (Shp2) has been implicated in various respiratory diseases; however, its role in PM2.5-induced epithelial barrier dysfunction remains unclear. Herein, we assessed the regulatory effects of Shp2 on PM2.5-mediated epithelial barrier function and tight junction (TJ) protein expression in both mice and human pulmonary epithelial (16HBE) cells. We observed that Shp2 levels were upregulated and claudin-4 levels were downregulated after PM2.5 stimulation in vivo and in vitro. Mice were exposed to PM2.5 to induce acute lung injury, and disrupted epithelial barrier function, with decreased transepithelial electrical resistance (TER) and increased paracellular flux that was observed in 16HBE cells. In contrast, the selective inhibition or knockdown of Shp2 retained airway epithelial barrier function and reversed claudin-4 downregulation that triggered by PM2.5, and these effects may occur through the ERK1/2 MAPK signaling pathway. These data highlight an important role of Shp2 in PM2.5-induced airway epithelial barrier dysfunction and suggest a possible new course of therapy for PM2.5-induced respiratory diseases.
Collapse
Affiliation(s)
- Youting Zhang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Likang Zhang
- Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wanwan Chen
- Department of Pathology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuanyuan Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoming Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaoyao Dong
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xixi Lin
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
17
|
Tripathi A, Kumar B, Sagi SSK. Hypoxia-mediated alterations in pulmonary surfactant protein expressions: Beneficial effects of quercetin prophylaxis. Respir Physiol Neurobiol 2021; 291:103695. [PMID: 34052411 DOI: 10.1016/j.resp.2021.103695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/26/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
We have compared the prophylactic efficacies of quercetin and salbutamol in preventing pulmonary surfactants oxidation under hypoxia. Male SD rats supplemented orally with quercetin (50 mg/Kg BW) and salbutamol (2 mg/Kg BW) were exposed to hypobaric hypoxia (7,620 m for 6 h). Hypoxia-mediated elevation in oxidative stress, inflammation, and extravasations of LDH & albumin content in BALF of rats were assessed. Western blotting and mRNA studies determined the differential expressions of Nrf-2, HO-1, and associated surfactant proteins (SP-A, SP-B, SP-C, & SP-D) in rat lungs. Later, the lung configuration under hypoxia was assessed histopathologically. Quercetin and salbutamol pretreatment considerably restored the expressions of Nrf-2, HO-1, and surfactant proteins to normal by attenuating the increase in oxidative stress, inflammation, and extravasations of plasma proteins in the animals under hypoxia. The histopathology has also evidenced the protective effect of quercetin in retaining normal lung architecture under hypoxia over salbutamol. The present study indicates the effectiveness of quercetin prophylaxis in preventing pulmonary surfactants oxidation under hypoxia over salbutamol.
Collapse
Affiliation(s)
- Ankit Tripathi
- Nutrition Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Bhuvnesh Kumar
- Nutrition Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Sarada S K Sagi
- Nutrition Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
18
|
Pao HP, Liao WI, Tang SE, Wu SY, Huang KL, Chu SJ. Suppression of Endoplasmic Reticulum Stress by 4-PBA Protects Against Hyperoxia-Induced Acute Lung Injury via Up-Regulating Claudin-4 Expression. Front Immunol 2021; 12:674316. [PMID: 34122432 PMCID: PMC8194262 DOI: 10.3389/fimmu.2021.674316] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
Endoplasmic reticulum (ER) stress that disrupts ER function can occur in response to a wide variety of cellular stress factors leads to the accumulation of unfolded and misfolded proteins in the ER. Many studies have shown that ER stress amplified inflammatory reactions and was involved in various inflammatory diseases. However, little is known regarding the role of ER stress in hyperoxia-induced acute lung injury (HALI). This study investigated the influence of ER stress inhibitor, 4-phenyl butyric acid (4-PBA), in mice with HALI. Treatment with 4-PBA in the hyperoxia groups significantly prolonged the survival, decreased lung edema, and reduced the levels of inflammatory mediators, lactate dehydrogenase, and protein in bronchoalveolar lavage fluid, and increased claudin-4 protein expression in lung tissue. Moreover, 4-PBA reduced the ER stress-related protein expression, NF-κB activation, and apoptosis in the lung tissue. In in vitro study, 4-PBA also exerted a similar effect in hyperoxia-exposed mouse lung epithelial cells (MLE-12). However, when claudin-4 siRNA was administrated in mice and MLE-12 cells, the protective effect of 4-PBA was abrogated. These results suggested that 4-PBA protected against hyperoxia-induced ALI via enhancing claudin-4 expression.
Collapse
Affiliation(s)
- Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
19
|
Tripathi A, Hazari PP, Mishra AK, Kumar B, Sagi SSK. Quercetin: a savior of alveolar barrier integrity under hypoxic microenvironment. Tissue Barriers 2021; 9:1883963. [PMID: 33632082 DOI: 10.1080/21688370.2021.1883963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
High altitude pulmonary edema (HAPE) is generally characterized by the loss of alveolar epithelial barrier integrity. The current study was undertaken to assess the noninvasive approaches of HAPE diagnosis and to evaluate the prophylactic potential of quercetin in preventing alveolar junction impairments. Male SD rats fed with quercetin 1 h prior to hypoxia (7,620 m, for 6 h) were selected. PET/CT imaging was performed to visualize the lung uptake of 18F-FDG in animals under hypoxia. Further, oxidant status, catalase activity, hematological & blood gas parameters were evaluated. Moreover, tight junction (TJ) proteins (ZO-1, JAM-C, Claudin-4, and occludin) expression analysis was accomplished using immune-blotting. The structural differences in lung epithelia were noted by TEM imaging. Quercetin prophylaxis has significantly reduced the FDG uptake in rat lungs under hypoxia. It has also dramatically alleviated the protein oxidation followed by an elevation in catalase activity in the lungs under hypoxia. The TJ protein expression in the lungs has also been restored to normal upon quercetin pre-treatment. Concomitantly, the quercetin preconditioning has elicited the stable blood gas and hematological parameters under hypoxia. The observations from TEM imaging have also implicated the normal lung epithelial structures in the quercetin pretreated animals under hypoxia. Quercetin prophylaxis has significantly restored alveolar epithelium integrity by abating oxidative stress in the lungs under hypoxia.Abbreviations: CT- Computed Tomography18F-FDG- Fluorodeoxyglucose (18FHAPE- High Altitude Pulmonary EdemaHb- HemoglobinHCT- HematocritHCO3- BicarbonateJAM- Junctional Adhesion MoleculeKBq- Killo BecquerelPaO2- Partial pressure of arterial oxygenPaCO2- Partial pressure of arterial carbon di-oxidePET- Positron Emission TomographyRBC- Red Blood CorpusclesSD- Sprague DawleyTJ- Tight JunctionsTEM- Transmission Electron MicroscopyWBC- White Blood CorpusclesZO- Zona Occludin.
Collapse
Affiliation(s)
- Ankit Tripathi
- Hematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur Delhi-India
| | - Puja P Hazari
- Molecular Imaging and Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi-India
| | - Anil K Mishra
- Molecular Imaging and Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi-India
| | - Bhuvnesh Kumar
- Hematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur Delhi-India
| | - Sarada S K Sagi
- Hematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur Delhi-India
| |
Collapse
|
20
|
Li J, Qi Z, Li D, Huang X, Qi B, Feng J, Qu J, Wang X. Alveolar epithelial glycocalyx shedding aggravates the epithelial barrier and disrupts epithelial tight junctions in acute respiratory distress syndrome. Biomed Pharmacother 2021; 133:111026. [PMID: 33378942 PMCID: PMC7685063 DOI: 10.1016/j.biopha.2020.111026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/08/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023] Open
Abstract
The main pathophysiological mechanism of acute respiratory distress syndrome (ARDS) invovles the increase in alveolar barrier permeability that is primarily caused by epithelial glycocalyx and tight junction (TJ) protein destruction. This study was performed to explore the effects of the alveolar epithelial glycocalyx on the epithelial barrier, specifically on TJ proteins, in ARDS. We used C57BL/6 mice and human lung epithelial cell models of lipopolysaccharide (LPS)-induced ARDS. Changes in alveolar permeability were evaluated via pulmonary histopathology analysis and by measuring the wet/dry weight ratio of the lungs. Degradation of heparan sulfate (HS), an important component of the epithelial glycocalyx, and alterations in levels of the epithelial TJ proteins (occludin, zonula occludens 1, and claudin 4) were assessed via ELISA, immunofluorescence analysis, and western blotting analysis. Real-time quantitative polymerase chain reaction was used to detect the mRNA of the TJ protein. Changes in glycocalyx and TJ ultrastructures in alveolar epithelial cells were evaluated through electron microscopy. In vivo and in vitro, LPS increased the alveolar permeability and led to HS degradation and TJ damage. After LPS stimulation, the expression of the HS-degrading enzyme heparanase (HPA) in the alveolar epithelial cells was increased. The HPA inhibitor N-desulfated/re-N-acetylated heparin alleviated LPS-induced HS degradation and reduced TJ damage. In vitro, recombinant HPA reduced the expression of the TJ protein zonula occludens-1 (ZO-1) and inhibited its mRNA expression in the alveolar epithelial cells. Taken together, our results demonstrate that shedding of the alveolar epithelial glycocalyx aggravates the epithelial barrier and damages epithelial TJ proteins in ARDS, with the underlying mechanism involving the effect of HPA on ZO-1.
Collapse
Affiliation(s)
- Jun Li
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Deparetment of Pulmonary and Critical Care Medicine, Yantai Affiliated Hospital of Binzhou Medical University, YanTai, Shandong, 264100, China
| | - Zhijiang Qi
- Deparetment of Pulmonary and Critical Care Medicine, Yantai Affiliated Hospital of Binzhou Medical University, YanTai, Shandong, 264100, China
| | - Dongxiao Li
- Department of Pulmonary and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Xiao Huang
- Department of Pulmonary and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Boyang Qi
- Department of Pulmonary and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Jiali Feng
- Department of Pulmonary and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Jianyu Qu
- Department of Pulmonary and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Xiaozhi Wang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Department of Pulmonary and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China.
| |
Collapse
|
21
|
Niroomand A, Lindstedt S. Is Claudin 4 a Player to Be Reckoned with or Not in the Context of Lung Transplantation? J INVEST SURG 2020; 35:201-202. [PMID: 33021126 DOI: 10.1080/08941939.2020.1825886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Anna Niroomand
- Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
22
|
Kodera Y, Kohno T, Konno T, Arai W, Tsujiwaki M, Shindo Y, Chiba H, Miyakawa M, Tanaka H, Sakuma Y, Watanabe A, Takahashi H, Kojima T. HMGB1 enhances epithelial permeability via p63/TGF-β signaling in lung and terminal bronchial epithelial cells. Tissue Barriers 2020; 8:1805997. [PMID: 32857676 PMCID: PMC7714505 DOI: 10.1080/21688370.2020.1805997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/28/2022] Open
Abstract
High mobility group box 1 (HMGB1) is involved in the induction of airway inflammation and injury in patients with chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). HMGB1 increased by transforming growth factor-β1 (TGF-β1), impairs airway epithelial barrier function in the lung. In the present study, to investigate how HMGB1 affects the barrier of normal human lung epithelial (HLE) cells, monolayer cells (2D culture) and bronchial-like spheroid cells (2.5 D Matrigel culture), which have lumen formation, were pretreated with TGF-β type I receptor kinase inhibitor EW-7197 before treatment with HMGB1. In 2D culture, treatment with HMGB1 decreased expression of angulin-1/LSR, TRIC and CLDN-1, -4, -7 and increased that of CLDN-2. Pretreatment with EW-7197 prevented the changes of all tight junction molecules induced by HMGB1. In 2.5D Matrigel culture, treatment with HMGB1 induced permeability of FITC-dextran (FD-4) into the lumen, whereas pretreatment with EW-7197 prevented the hyperpermeability of FD-4 into the lumen caused by HMGB1. In 2.5D Matrigel culture, knockdown of transcription factor p63 prevented the hyperpermeability induced by HMGB1 as well as pretreatment with EW-7197. In the 2D culture of HLE cells with HMGB1, knockdown of p63 increased the level of angulin-1/LSR and CLDN-4, while pretreatment with EW-7197 enhanced the increase of CLDN-4 induced by knockdown of p63. Immunohistochemical analysis of IPF, CLDN-2, HMGB1 and p63 revealed that their levels were higher in the regenerative epithelium of the terminal bronchial region than in normal epithelium. HMGB1 induces epithelial permeability of HLE cells via p63/TGF-β signaling in normal lung and IPF.
Collapse
Affiliation(s)
- Yuki Kodera
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Wataru Arai
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Thoracic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsuhiro Tsujiwaki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuma Shindo
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Thoracic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Maki Miyakawa
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- IBD Center, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Hiroki Tanaka
- IBD Center, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Yuji Sakuma
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Watanabe
- Department of Thoracic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
23
|
Luna-Flores A, Olmos-Zuñiga JR, Jasso-Victoria R, Gaxiola-Gaxiola M, Aguirre-Pérez T, Ruiz V, García-Torrentera R, Silva-Martínez M, Zenteno E, Gutierrez-Ospina G, Santillan-Doherty P. Expression of Claudin-4 in Lung Ischemia-Reperfusion Injury in Experimental Lung Transplantation. J INVEST SURG 2020; 35:191-200. [PMID: 32900258 DOI: 10.1080/08941939.2020.1815253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To assess the presence of CLDN4 in bronchoalveolar lavage fluid (BALF) and pulmonary tissue as an early indicator of LIRI and its relationship with changes in pulmonary physiology, edema formation and histology in an experimental porcine model of LTx with CIT of 50 min or 6 h. METHODS In 12 pigs, LIRI was produced by: group I (n = 6) LTx with 50 min of CIT (LTx-50 min-CIT); and group II (n = 6) LTx with 6 h of CIT (LTx-6h-CIT). The lung function, edema formation, macroscopic and microscopic changes were assessed. CLDN4 expression in BALF and pulmonary tissue were determined. RESULTS Both groups presented similar clinical, edema, and histological damage, as well as similar expression of CLDN4 in BALF and tissue (p > 0.05, RM-ANOVA). CONCLUSION CLDN4 expressed in BALF and the pulmonary tissue during the first 5 h within 72 h of the PGD window are not associated by the deterioration of lung function, edema and lung histological injury, in LTx with CIT 50 min or 6 h, CLDN4 does not seem to be a valuable indicator of LIRI.
Collapse
Affiliation(s)
- Antonia Luna-Flores
- Lung Transplantation Research Unit, Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - J Raúl Olmos-Zuñiga
- Lung Transplantation Research Unit, Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Rogelio Jasso-Victoria
- Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Miguel Gaxiola-Gaxiola
- Department of Morphology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Teresa Aguirre-Pérez
- Bronchoscopy and Endoscopy Service, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Victor Ruiz
- Molecular Biology Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Rogelio García-Torrentera
- Respiratory Emergency Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Mariana Silva-Martínez
- Lung Transplantation Research Unit, Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Edgar Zenteno
- Department of Biochemistry, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Gabriel Gutierrez-Ospina
- Department of Cell Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Patricio Santillan-Doherty
- Medical Direction, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
24
|
Zhang H, Cui Y, Zhou Z, Ding Y, Nie H. Alveolar Type 2 Epithelial Cells as Potential Therapeutics for Acute Lung Injury/Acute Respiratory Distress Syndrome. Curr Pharm Des 2020; 25:4877-4882. [PMID: 31801451 DOI: 10.2174/1381612825666191204092456] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
Acute lung injury/acute respiratory distress syndrome is a common clinical illness with high morbidity and mortality, which is still one of the medical problems urgently needed to be solved. Alveolar type 2 epithelial cells are an important component of lung epithelial cells and as a kind of stem cells, they can proliferate and differentiate into alveolar type 1 epithelial cells, thus contributing to lung epithelial repairment. In addition, they synthesize and secrete all components of the surfactant that regulates alveolar surface tension in the lungs. Moreover, alveolar type 2 epithelial cells play an active role in enhancing alveolar fluid clearance and reducing lung inflammation. In recent years, as more advanced approaches appear in the field of stem and progenitor cells in the lung, many preclinical studies have shown that the cell therapy of alveolar type 2 epithelial cells has great potential effects for acute lung injury/acute respiratory distress syndrome. We reviewed the recent progress on the mechanisms of alveolar type 2 epithelial cells involved in the damaged lung repairment, aiming to explore the possible therapeutic targets in acute lung injury/acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Honglei Zhang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yong Cui
- Department of Anesthesiology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyu Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
25
|
Li J, Jie X, Liang X, Chen Z, Xie P, Pan X, Zhou B, Li J. Sinensetin suppresses influenza a virus-triggered inflammation through inhibition of NF-κB and MAPKs signalings. BMC Complement Med Ther 2020; 20:135. [PMID: 32370749 PMCID: PMC7200050 DOI: 10.1186/s12906-020-02918-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background Human respiratory system infected with influenza A virus (IAV) elicited a robust pro-inflammatory response that resulted in severe illness and even death. Currently, limited immunomodulator is available to counteract IAV-associated pneumonia in the clinic. Sinensetin, a polymethoxylated flavone with five methoxy groups, has been found to possess anti-agiogenesis, anti-inflammatory and anti-diabetic activities. However, the effects of sinensetin on IAV-triggered pro-inflammatory response remain unclear. In the present study, the anti-inflammatory effects and corresponding possible mechanism of sinensetin in IAV-infected A549 cells were subjected to investigations. Methods The cytotoxic effects of sinensetin towards A549 cells was detected by MTT and LDH assays. The antiviral activity of sinensetin against influenza A virus was assayed in A549 cells with an engineered replication-competent influenza A virus carrying Gaussia luciferase reporter gene infection. The effect of sinensetin on influenza A virus-triggered inflammatory reaction was determined by qRT-PCR, Luminex assays, ELISA and Western blot. Results Our results showed that sinensetin did not exhibit antiviral activity against A/PR/8/34 (H1N1). Meanwhile, sinensetin treatment significantly decreased IAV-induced expression of pro-inflammatory mediators at mRNA and protein levels, including IL-6, TNF-α, IP-10, IL-8 and MCP-1. Additionally, levels of cyclooxygenase (COX)-2 and the downstream product prostaglandin E2 (PGE2) up-regulated by IAV infection were dramatically suppressed by sinensetin. The mechanistic investigation revealed that sinensetin treatment suppressed the NF-κB transcriptional activity using the NF-κB reporter stable HEK293 cell line stimulated with TNF-α (20 ng/mL) or influenza H1N1 virus. Furthermore, sinensetin abrogated influenza H1N1 virus-induced activation of NF-κB, ERK1/2 MAPK and p38 MAPK signalings. Conclusion Collectively, our results indicated that sinensetin has potential capacity to attenuate IAV-triggered pro-inflammatory response via inactivation of NF-κB, ERK1/2 MAPK and p38 MAPK signalings, which implied that sinensetin may be a promising candidate drug for influenza H1N1 virus infection therapeutics.
Collapse
Affiliation(s)
- Jiashun Li
- Department of Respiratory, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Huadu, Guangzhou, Guangdong, 510800, P.R. China
| | - Xiang Jie
- Huizhou third people's hospital, Guangzhou Medical University, Guangdong, 516002, China
| | - Xiaoli Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, P.R. China
| | - Ziyu Chen
- Institute of Respiratory Diseases, Department of Respiratory, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Peifang Xie
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, P.R. China
| | - Xiping Pan
- Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P.R. China
| | - Beixian Zhou
- Department of Pharmacy, The People's hospital of Gaozhou, Gaozhou, 525200, Guangdong, China.
| | - Jing Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, P.R. China.
| |
Collapse
|
26
|
Zou J, Li Y, Yu J, Dong L, Husain AN, Shen L, Weber CR. Idiopathic pulmonary fibrosis is associated with tight junction protein alterations. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183205. [DOI: 10.1016/j.bbamem.2020.183205] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 01/07/2020] [Accepted: 01/18/2020] [Indexed: 02/07/2023]
|
27
|
Abstract
The pulmonary blood-gas barrier represents a remarkable feat of engineering. It achieves the exquisite thinness needed for gas exchange by diffusion, the strength to withstand the stresses and strains of repetitive and changing ventilation, and the ability to actively maintain itself under varied demands. Understanding the design principles of this barrier is essential to understanding a variety of lung diseases, and to successfully regenerating or artificially recapitulating the barrier ex vivo. Many classical studies helped to elucidate the unique structure and morphology of the mammalian blood-gas barrier, and ongoing investigations have helped to refine these descriptions and to understand the biological aspects of blood-gas barrier function and regulation. This article reviews the key features of the blood-gas barrier that enable achievement of the necessary design criteria and describes the mechanical environment to which the barrier is exposed. It then focuses on the biological and mechanical components of the barrier that preserve integrity during homeostasis, but which may be compromised in certain pathophysiological states, leading to disease. Finally, this article summarizes recent key advances in efforts to engineer the blood-gas barrier ex vivo, using the platforms of lung-on-a-chip and tissue-engineered whole lungs. © 2020 American Physiological Society. Compr Physiol 10:415-452, 2020.
Collapse
Affiliation(s)
- Katherine L. Leiby
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Micha Sam Brickman Raredon
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Laura E. Niklason
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Yale School of Medicine, Yale University, New Haven, Connecticut, USA
- Department of Anesthesiology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
28
|
The IL1β-HER2-CLDN18/CLDN4 axis mediates lung barrier damage in ARDS. Aging (Albany NY) 2020; 12:3249-3265. [PMID: 32065780 PMCID: PMC7066891 DOI: 10.18632/aging.102804] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/19/2020] [Indexed: 12/17/2022]
Abstract
Objective: The high mortality rate associated with acute respiratory distress syndrome (ARDS) is a major challenge for intensive care units. In the present study, we applied bioinformatics and animal models to identify core genes and potential corresponding pathways in ARDS. Results: Using bioinformatics analysis, IL-1β was identified as the core gene of ARDS. Cell experiments showed that up-regulation of IL-1β downregulates claudin18 to promote lung barrier function damage by regulating the IL-1β-HER2/HER3 axis, further promoting the development of ARDS. This was validated in the animal models. Conclusion: IL-1β promotes the development of ARDS by regulating the IL-1β-HER2/HER3 axis. These findings deepen the understanding of the pathological mechanisms of ARDS. Methods: Transcription data sets related to ARDS were subjected to differential expression gene analysis, functional enrichment analysis, and receiver operating characteristic curve analysis and, so as to identify core genes in ARDS. Cell experiments were used to further explore the effects of core genes on lung barrier function damage. Animal models were applied to validate the effects of core gene in mediating biological signal pathways in ARDS.
Collapse
|
29
|
Johnson AN, Harkema JR, Nelson AJ, Dickinson JD, Kalil J, Duryee MJ, Thiele GM, Kumar B, Singh AB, Gaurav R, Glover SC, Tang Y, Romberger DJ, Kielian T, Poole JA. MyD88 regulates a prolonged adaptation response to environmental dust exposure-induced lung disease. Respir Res 2020; 21:97. [PMID: 32321514 PMCID: PMC7178993 DOI: 10.1186/s12931-020-01362-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Environmental organic dust exposures enriched in Toll-like receptor (TLR) agonists can reduce allergic asthma development but are associated with occupational asthma and chronic bronchitis. The TLR adaptor protein myeloid differentiation factor88 (MyD88) is fundamental in regulating acute inflammatory responses to organic dust extract (ODE), yet its role in repetitive exposures is unknown and could inform future strategies. METHODS Wild-type (WT) and MyD88 knockout (KO) mice were exposed intranasally to ODE or saline daily for 3 weeks (repetitive exposure). Repetitively exposed animals were also subsequently rested with no treatments for 4 weeks followed by single rechallenge with saline/ODE. RESULTS Repetitive ODE exposure induced neutrophil influx and release of pro-inflammatory cytokines and chemokines were profoundly reduced in MyD88 KO mice. In comparison, ODE-induced cellular aggregates, B cells, mast cell infiltrates and serum IgE levels remained elevated in KO mice and mucous cell metaplasia was increased. Expression of ODE-induced tight junction protein(s) was also MyD88-dependent. Following recovery and then rechallenge with ODE, inflammatory mediators, but not neutrophil influx, was reduced in WT mice pretreated with ODE coincident with increased expression of IL-33 and IL-10, suggesting an adaptation response. Repetitively exposed MyD88 KO mice lacked inflammatory responsiveness upon ODE rechallenge. CONCLUSIONS MyD88 is essential in mediating the classic airway inflammatory response to repetitive ODE, but targeting MyD88 does not reduce mucous cell metaplasia, lymphocyte influx, or IgE responsiveness. TLR-enriched dust exposures induce a prolonged adaptation response that is largely MyD88-independent. These findings demonstrate the complex role of MyD88-dependent signaling during acute vs. chronic organic dust exposures.
Collapse
Affiliation(s)
- Amber N. Johnson
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA
| | - Jack R. Harkema
- grid.17088.360000 0001 2150 1785Pathobiology & Diagnostic Investigation, Institute for Integrative Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI USA
| | - Amy J. Nelson
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA
| | - John D. Dickinson
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA
| | - Julianna Kalil
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA
| | - Michael J. Duryee
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA ,Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE USA
| | - Geoffrey M. Thiele
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA ,Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE USA
| | - Balawant Kumar
- grid.266813.80000 0001 0666 4105Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE USA
| | - Amar B. Singh
- Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE USA ,grid.266813.80000 0001 0666 4105Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE USA
| | - Rohit Gaurav
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA
| | - Sarah C. Glover
- grid.410721.10000 0004 1937 0407Department of Medicine, University of Mississippi Medical Center, Jackson, MS USA
| | - Ying Tang
- grid.15276.370000 0004 1936 8091Department of Medicine, University of Florida, Gainesville, FL USA
| | - Debra J. Romberger
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA ,Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE USA
| | - Tammy Kielian
- grid.266813.80000 0001 0666 4105Department of Microbiology and Pathology, University of Nebraska Medical Center, Omaha, NE USA
| | - Jill A. Poole
- grid.266813.80000 0001 0666 4105Department of Internal Medicine, University of Nebraska Medical Center, 985990 Nebraska Medical Center, Omaha, NE 68198-5990 USA
| |
Collapse
|
30
|
Hebert KD, Mclaughlin N, Galeas-Pena M, Zhang Z, Eddens T, Govero A, Pilewski JM, Kolls JK, Pociask DA. Targeting the IL-22/IL-22BP axis enhances tight junctions and reduces inflammation during influenza infection. Mucosal Immunol 2020; 13:64-74. [PMID: 31597930 PMCID: PMC6917921 DOI: 10.1038/s41385-019-0206-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 08/20/2019] [Accepted: 09/02/2019] [Indexed: 02/04/2023]
Abstract
The seasonal burden of influenza coupled with the pandemic outbreaks of more pathogenic strains underscore a critical need to understand the pathophysiology of influenza injury in the lung. Interleukin-22 (IL-22) is a promising cytokine that is critical in protecting the lung during infection. This cytokine is strongly regulated by the soluble receptor IL-22-binding protein (IL-22BP), which is constitutively expressed in the lungs where it inhibits IL-22 activity. The IL-22/IL-22BP axis is thought to prevent chronic exposure of epithelial cells to IL-22. However, the importance of this axis is not understood during an infection such as influenza. Here we demonstrate through the use of IL-22BP-knockout mice (il-22ra2-/-) that a pro-IL-22 environment reduces pulmonary inflammation during H1N1 (PR8/34 H1N1) infection and protects the lung by promoting tight junction formation. We confirmed these results in normal human bronchial epithelial cells in vitro demonstrating improved membrane resistance and induction of the tight junction proteins Cldn4, Tjp1, and Tjp2. Importantly, we show that administering recombinant IL-22 in vivo reduces inflammation and fluid leak into the lung. Taken together, our results demonstrate the IL-22/IL-22BP axis is a potential targetable pathway for reducing influenza-induced pneumonia.
Collapse
Affiliation(s)
- K D Hebert
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - N Mclaughlin
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - M Galeas-Pena
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Z Zhang
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - T Eddens
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, 15224, USA
| | - A Govero
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - J M Pilewski
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - J K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - D A Pociask
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
31
|
M T, T A, B S, Ak G, Sks S. Curcumin prophylaxis refurbishes alveolar epithelial barrier integrity and alveolar fluid clearance under hypoxia. Respir Physiol Neurobiol 2019; 274:103336. [PMID: 31778793 DOI: 10.1016/j.resp.2019.103336] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022]
Abstract
We have studied the prophylactic efficacy of curcumin to ameliorate the impairment of tight junction protein integrity and fluid clearance in lungs of rats under hypoxia. A549 cells wereexposed to 3 % O2 for 1 h, 3 h, 6 h, 12 h, 24 h and 48 h and rats were exposed to 7620 m for 6 h. NF-κB, Hif-1α and their related genes, tight junction protein (TJ) (ZO-1, JAM-C, claudin-4 and claudin-5, claudin-18) expressions were determined in A549 cells and lungs of rats by western blotting, ELISA and their activity by reporter gene assay, siRNAp65 knock out. Tissue specific localization of tight junction protein was determined by immunohistochemistry and immunoflorescence. Further transmission electron microscopy (TEM) was used to visualize the TJ structures between pulmonary epithelial cells. Blood gas and hematological parameters were also assessed. Later we checked, whether prior treatment with curcumin can restore the altered alveolar epithelial barrier integrity that is compromised through inflammatory mediators under hypoxia, A549 cells were pre-treated (1 h) with 10 μM curcumin and rats with 50 mg curcumin/kg BW and exposed to hypoxia. Curcumin pre-treatment both in vitro and in vivo showed significant changes in TJ protein integrity, attenuated NF-κB activity with reduced expression of its regulatory genes in lung tissues, serum and bronchoalveolar lavage fluid (BALF) along with stabilized HIF-1α levels under hypoxia. NF-κB inhibitors MG132, SN50 or siRNA mediated p65 knock down significantly reduced the dextran FITC influx into the lungs. The present study indicates that, curcumin prophylaxis augments alveolar epithelial barrier integrity and alveolar fluid clearance under hypoxia.
Collapse
Affiliation(s)
- Titto M
- Haematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India.
| | - Ankit T
- Haematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India.
| | - Saumya B
- Haematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India.
| | - Gausal Ak
- Haematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India.
| | - Sarada Sks
- Haematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India.
| |
Collapse
|
32
|
Dai YL, Wu CP, Yang GG, Chang H, Peng CK, Huang KL. Adaptive Support Ventilation Attenuates Ventilator Induced Lung Injury: Human and Animal Study. Int J Mol Sci 2019; 20:ijms20235848. [PMID: 31766467 PMCID: PMC6929029 DOI: 10.3390/ijms20235848] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 01/10/2023] Open
Abstract
Adaptive support ventilation (ASV) is a closed-loop ventilation, which can make automatic adjustments in tidal volume (VT) and respiratory rate based on the minimal work of breathing. The purpose of this research was to study whether ASV can provide a protective ventilation pattern to decrease the risk of ventilator-induced lung injury in patients of acute respiratory distress syndrome (ARDS). In the clinical study, 15 ARDS patients were randomly allocated to an ASV group or a pressure-control ventilation (PCV) group. There was no significant difference in the mortality rate and respiratory parameters between these two groups, suggesting the feasible use of ASV in ARDS. In animal experiments of 18 piglets, the ASV group had a lower alveolar strain compared with the volume-control ventilation (VCV) group. The ASV group exhibited less lung injury and greater alveolar fluid clearance compared with the VCV group. Tissue analysis showed lower expression of matrix metalloproteinase 9 and higher expression of claudin-4 and occludin in the ASV group than in the VCV group. In conclusion, the ASV mode is capable of providing ventilation pattern fitting into the lung-protecting strategy; this study suggests that ASV mode may effectively reduce the risk or severity of ventilator-associated lung injury in animal models.
Collapse
Affiliation(s)
- Yu-Ling Dai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Division of Pulmonary and Critical Care Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Taiwan Landseed Hospital, Tao-Yuan 32449, Taiwan;
| | - Gee-Gwo Yang
- Division of Chest Medicine, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan;
| | - Hung Chang
- Department of Physiology, National Defense Medical Center, Taipei 114, Taiwan;
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: (C.-K.P.); (K.-L.H.); Tel.: +886-2-8792-3311 (ext. 13482) (C.-K.P.); +886-2-8792-3311 (ext. 12464) (K.-L.H)
| | - Kun-Lun Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Division of Pulmonary and Critical Care Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: (C.-K.P.); (K.-L.H.); Tel.: +886-2-8792-3311 (ext. 13482) (C.-K.P.); +886-2-8792-3311 (ext. 12464) (K.-L.H)
| |
Collapse
|
33
|
Luo Z, Li M, Wu Y, Meng Z, Martin L, Zhang L, Ogunrinde E, Zhou Z, Qin S, Wan Z, Westerink MAJ, Warth S, Liu H, Jin P, Stroncek D, Li QZ, Wang E, Wu X, Heath SL, Li Z, Alekseyenko AV, Jiang W. Systemic translocation of Staphylococcus drives autoantibody production in HIV disease. MICROBIOME 2019; 7:25. [PMID: 30764863 PMCID: PMC6376754 DOI: 10.1186/s40168-019-0646-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 02/05/2019] [Indexed: 05/12/2023]
Abstract
BACKGROUND Increased autoreactive antibodies have been reported in HIV disease; however, the mechanism accounting for autoantibody induction in HIV remains unknown. RESULTS Herein, we show that seasonal influenza vaccination induces autoantibody production (e.g., IgG anti-nuclear antibody (ANA) and anti-double-stranded DNA antibody (anti-dsDNA)) in some viral-suppressed antiretroviral therapy (ART)-treated HIV+ subjects, but not in healthy controls. These autoantibodies were not derived from antigen-specific B cells but from activated "bystander" B cells analyzed by single-cell assay and by study of purified polyclonal ANAs from plasma. To explore the mechanism of autoantibody generation in HIV+ subjects, plasma level of microbial products, gene expression profile of B cells, and B cell receptor (BCR) repertoires were analyzed. We found that autoantibody production was associated with increased plasma level of microbial translocation; the patients with high autoantibodies had skewed B cell repertoires and upregulation of genes related to innate immune activation in response to microbial translocation. By analyzing circulating microbial 16S rDNA in plasma, the relative abundance of Staphylococcus was found to be associated with autoantibody production in HIV+ subjects. Finally, we found that injection of heat-killed Staphylococcus aureus promoted germinal center B cell responses and autoantibody production in mice, consistent with the notion that autoantibody production in HIV+ patients is triggered by microbial products. CONCLUSIONS Our results showed that translocation of Staphylococcus can promote B cell activation through enhancing germinal center response and induces autoantibody production. It uncovers a potential mechanism linking microbial translocation and autoimmunity in HIV+ disease and provides a strong rationale for targeting Staphylococcus to prevent autoantibody production.
Collapse
Affiliation(s)
- Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Min Li
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Zhefeng Meng
- Department of Gastroenterology, Oncology Bioinformatics Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Lisa Martin
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Lumin Zhang
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Elizabeth Ogunrinde
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Zejun Zhou
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Shenghui Qin
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Zhuang Wan
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Maria Anna Julia Westerink
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Stephanie Warth
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Hui Liu
- Cell Processing Section (CPS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, 20892, USA
| | - Ping Jin
- Cell Processing Section (CPS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, 20892, USA
| | - David Stroncek
- Cell Processing Section (CPS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, 20892, USA
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Ena Wang
- Sidra Medical and Research Center, Doha, Qatar
| | - Xueling Wu
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY, 10016, USA
| | - Sonya L Heath
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Alexander V Alekseyenko
- Program for Human Microbiome Research, Biomedical Informatics Center, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA.
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
34
|
Tan HTT, Hagner S, Ruchti F, Radzikowska U, Tan G, Altunbulakli C, Eljaszewicz A, Moniuszko M, Akdis M, Akdis CA, Garn H, Sokolowska M. Tight junction, mucin, and inflammasome-related molecules are differentially expressed in eosinophilic, mixed, and neutrophilic experimental asthma in mice. Allergy 2019; 74:294-307. [PMID: 30267575 DOI: 10.1111/all.13619] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/16/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Asthma is a chronic respiratory disease with marked clinical and pathophysiological heterogeneity. Specific pathways are thought to be involved in the pathomechanisms of different inflammatory phenotypes of asthma; however, direct in vivo comparison has not been performed. METHODS We developed mouse models representing three different phenotypes of allergic airway inflammation-eosinophilic, mixed, and neutrophilic asthma via different methods of house dust mite sensitization and challenge. Transcriptomic analysis of the lungs, followed by the RT-PCR, western blot, and confocal microscopy, was performed. Primary human bronchial epithelial cells cultured in air-liquid interface were used to study the mechanisms revealed in the in vivo models. RESULTS By whole-genome transcriptome profiling of the lung, we found that airway tight junction (TJ), mucin, and inflammasome-related genes are differentially expressed in these distinct phenotypes. Further analysis of proteins from these families revealed that Zo-1 and Cldn18 were downregulated in all phenotypes, while increased Cldn4 expression was characteristic for neutrophilic airway inflammation. Mucins Clca1 (Gob5) and Muc5ac were upregulated in eosinophilic and even more in neutrophilic phenotype. Increased expression of inflammasome-related molecules such as Nlrp3, Nlrc4, Casp-1, and IL-1β was characteristic for neutrophilic asthma. In addition, we showed that inflammasome/Th17/neutrophilic axis cytokine-IL-1β-may transiently impair epithelial barrier function, while IL-1β and IL-17 increase mucin expressions in primary human bronchial epithelial cells. CONCLUSION Our findings suggest that differential expression of TJ, mucin, and inflammasome-related molecules in distinct inflammatory phenotypes of asthma may be linked to pathophysiology and might reflect the differences observed in the clinic.
Collapse
Affiliation(s)
- Hern-Tze Tina Tan
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
- Department of Immunology; School of Medical Sciences; Universiti Sains Malaysia; Kubang Kerian Malaysia
| | - Stefanie Hagner
- Institute of Laboratory Medicine and Pathobiochemistry; Molecular Diagnostics; Marburg Germany
- Member of the German Center for Lung Research; Marburg Germany
| | - Fiorella Ruchti
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
- Department of Regenerative Medicine and Immune Regulation; Medical University of Bialystok; Bialystok Poland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
- Functional Genomics Center Zurich; ETH Zurich/University of Zurich; Zurich Switzerland
| | - Can Altunbulakli
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
| | - Andrzej Eljaszewicz
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
- Department of Regenerative Medicine and Immune Regulation; Medical University of Bialystok; Bialystok Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation; Medical University of Bialystok; Bialystok Poland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry; Molecular Diagnostics; Marburg Germany
- Member of the German Center for Lung Research; Marburg Germany
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
- Christine Kühne-Center for Allergy Research and Education; Davos Switzerland
| |
Collapse
|
35
|
Dunberry-Poissant S, Gilbert K, Bouchard C, Baril F, Cardinal AM, L'Ecuyer S, Hylands M, Lamontagne F, Rousseau G, Charbonney E. Fluid sparing and norepinephrine use in a rat model of resuscitated haemorrhagic shock: end-organ impact. Intensive Care Med Exp 2018; 6:47. [PMID: 30421022 PMCID: PMC6232186 DOI: 10.1186/s40635-018-0212-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/29/2018] [Indexed: 12/02/2022] Open
Abstract
Background Haemostasis and correction of hypovolemia are the pillars of early haemorrhage shock (HS) management. Vasopressors, which are not recommended as first-line therapy, are an alternative to aggressive fluid resuscitation, but data informing the risks and benefits of vasopressor therapy as fluid-sparing strategy is lacking. We aimed to study its impact on end organs, in the setting of a haemodynamic response to the initial volume resuscitation. Methods Following controlled HS (60 min) induced by blood withdrawal, under anaesthesia and ventilation, male Wistar rats (N = 10 per group) were randomly assigned to (1) sham, (2) HS with fluid resuscitation only [FR] and (3) HS with fluid resuscitation to restore haemodynamic (MAP: mean arterial pressure) then norepinephrine [FR+NE]. After a reperfusion time (60 min) during which MAP was maintained with fluid or norepinephrine, equipment was removed and animals were observed for 24 h (N = 5) or 72 h (N = 5) before euthanasia. Besides haemodynamic parameters, physiological markers (creatinine, lactate, pH, PaO2) and one potential contributor to vasoplegia (xanthine oxidase activity) were measured. Apoptosis induction (caspase 3), tissue neutrophil infiltration (MPO: myeloperoxidase) and illustrative protein markers were measured in the lung (Claudin-4), kidney (KIM-1) and brain amygdala (Iba1). Results No difference was present in MAP levels during HS or reperfusion between the two resuscitation strategies. FR required significantly more fluid than FR+NE (183% vs 106% of bleed-out volume; p = 0.003), when plasma lactate increased similarly. Xanthine oxidase was equally activated in both HS groups. After FR+NE, creatinine peaked higher but was similar in all groups at later time points. FR+NE enhanced MPO in the lung, when Claudin-4 increased significantly after FR. In the brain amygdala, FR provoked more caspase 3 activity, MPO and microglial activation (Iba1 expression). Conclusion Organ resuscitation after controlled HS can be assured with lesser fluid administration followed by vasopressors administration, without signs of dysoxia or worse evolution. Limiting fluid administration could benefit the brain and seems not to have a negative impact on the lung or kidney.
Collapse
Affiliation(s)
- Sophie Dunberry-Poissant
- Département de Médecine, Université de Montréal, C.P. 6128 Succursale Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - Kim Gilbert
- Centre de Recherche Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 boul. Gouin Ouest, Montréal, QC, H4J 1C5, Canada
| | - Caroline Bouchard
- Centre de Recherche Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 boul. Gouin Ouest, Montréal, QC, H4J 1C5, Canada
| | - Frédérique Baril
- Université de Montréal, 2900 Edouard Montpetit Blvd, Montréal, QC, H3T 1J4, Canada
| | - Anne-Marie Cardinal
- Université de Montréal, 2900 Edouard Montpetit Blvd, Montréal, QC, H3T 1J4, Canada
| | - Sydnée L'Ecuyer
- Université de Montréal, 2900 Edouard Montpetit Blvd, Montréal, QC, H3T 1J4, Canada
| | - Mathieu Hylands
- Département de chirurgie, Université de Sherbrooke, 3001- 12e avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - François Lamontagne
- Centre de recherche du CHU de Sherbrooke, 3001- 12e avenue Nord, Sherbrooke, QC, J1H 5N4, Canada.,Department of Medicine, Université de Sherbrooke, 3001- 12e avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Guy Rousseau
- Centre de Recherche Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 boul. Gouin Ouest, Montréal, QC, H4J 1C5, Canada.,Département de pharmacologie et physiologie, Université de Montréal, C.P. 6128 Succursale Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - Emmanuel Charbonney
- Département de Médecine, Université de Montréal, C.P. 6128 Succursale Centre-ville, Montréal, QC, H3C 3J7, Canada. .,Centre de Recherche Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 boul. Gouin Ouest, Montréal, QC, H4J 1C5, Canada.
| |
Collapse
|
36
|
Chen H, Lu R, Zhang YG, Sun J. Vitamin D Receptor Deletion Leads to the Destruction of Tight and Adherens Junctions in Lungs. Tissue Barriers 2018; 6:1-13. [PMID: 30409076 DOI: 10.1080/21688370.2018.1540904] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vitamin D deficiency has been linked to various inflammatory diseases in lungs, including pneumonia, asthma and chronic obstructive pulmonary disease. However, the mechanisms by which vitamin D and vitamin D receptor reduce inflammation in lung diseases remain poorly understood. In this study, we investigated the expression and cell-specific distribution of tight and adherens junctions in the lungs of vitamin D receptor-deficient (VDR-/-) mice. Our results demonstrated that mRNA and protein levels of claudin-2, claudin-4 and claudin-12 were significantly decreased in the lungs of VDR-/- mice. Other tight and adherens junction proteins, such as ZO-1, occludin, claudin-10, β-catenin, and VE-cadherin, showed significant differences in expression in the lungs of VDR-/- and wild-type mice. These data suggest that altered expression of tight and adherens junction molecules, especially of claudin-2, -4, -10, -12, and -18, after chronic pneumonia caused by VDR deletion could increase lung permeability.Therefore, VDR may play an important role in maintaining pulmonary barrier integrity. Further studies should confirm whether vitamin D/VDR is beneficial for the prevention or treatment of lung diseases.
Collapse
Affiliation(s)
- Honglei Chen
- a Department of Biochemistry , Rush University , Chicago , IL , USA
| | - Rong Lu
- b Division of Gastroenterology and Hepatology, Department of Medicine , University of Illinois at Chicago , Chicago , IL , USA
| | - Yong-Guo Zhang
- b Division of Gastroenterology and Hepatology, Department of Medicine , University of Illinois at Chicago , Chicago , IL , USA
| | - Jun Sun
- b Division of Gastroenterology and Hepatology, Department of Medicine , University of Illinois at Chicago , Chicago , IL , USA
| |
Collapse
|
37
|
Dagenais A, Desjardins J, Shabbir W, Roy A, Filion D, Sauvé R, Berthiaume Y. Loss of barrier integrity in alveolar epithelial cells downregulates ENaC expression and activity via Ca 2+ and TRPV4 activation. Pflugers Arch 2018; 470:1615-1631. [PMID: 30088081 DOI: 10.1007/s00424-018-2182-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 06/14/2018] [Accepted: 07/09/2018] [Indexed: 01/08/2023]
Abstract
The epithelial Na channel (ENaC) plays an essential role in lung physiology by modulating the amount of liquid lining the respiratory epithelium. Here, we tested the effect of breaking alveolar epithelial cell barrier integrity on ENaC expression and function. We found that either mechanical wounding by scratching the monolayer or disruption of tight junction with EDTA induced a ~ 50% decrease of α,β and γENaC mRNA expression and an 80% reduction of ENaC short-circuit current (Isc) at 6 h. Scratching the cell monolayer generated a Ca2+ wave that spread from the margin of the scratch to distant cells. Pretreatment with BAPTA-AM, an intracellular Ca2+ chelator, abolished the effect of mechanical wounding and EDTA on αENaC mRNA expression, suggesting that [Ca2+]i is important for this modulation. We tested the hypothesis that a mechanosensitive channel such as TRPV4, a cationic channel known to increase [Ca2+]i, could mediate this effect. Activation of the channel with the TRPV4 specific agonist GSK-1016790A (GSK) decreased αENAC mRNA expression and almost completely abolished ENaC Isc. Pretreatment of alveolar epithelial cells with HC-067047 (HC0), a specific TRPV4 antagonist, reduced the extent of αENAC mRNA downregulation by mechanical wounding and EDTA. Altogether, our results suggest that mechanical stress induced by wounding or TRPV4-mediated loss of tight junction increases [Ca2+]i and elicits a Ca2+ wave that affects ENaC expression and function away from the site of injury. These data are important to better understand how Ca2+ signaling affects lung liquid clearance in injured lungs.
Collapse
Affiliation(s)
- André Dagenais
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada.
- Département de médecine, Université de Montréal, Montreal, Quebec, Canada.
| | - Julie Desjardins
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
| | - Waheed Shabbir
- Institute of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Antoine Roy
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
| | - Dominic Filion
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
| | - Rémy Sauvé
- Département de pharmacologie et physiologie, Université de Montréal, Montreal, Quebec, Canada
| | - Yves Berthiaume
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
- Département de médecine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
38
|
Magnani ND, Dada LA, Sznajder JI. Ubiquitin-proteasome signaling in lung injury. Transl Res 2018; 198:29-39. [PMID: 29752900 PMCID: PMC6986356 DOI: 10.1016/j.trsl.2018.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/15/2018] [Accepted: 04/16/2018] [Indexed: 12/21/2022]
Abstract
Cell homeostasis requires precise coordination of cellular proteins function. Ubiquitination is a post-translational modification that modulates protein half-life and function and is tightly regulated by ubiquitin E3 ligases and deubiquitinating enzymes. Lung injury can progress to acute respiratory distress syndrome that is characterized by an inflammatory response and disruption of the alveolocapillary barrier resulting in alveolar edema accumulation and hypoxemia. Ubiquitination plays an important role in the pathobiology of acute lung injury as it regulates the proteins modulating the alveolocapillary barrier and the inflammatory response. Better understanding of the signaling pathways regulated by ubiquitination may lead to novel therapeutic approaches by targeting specific elements of the ubiquitination pathways.
Collapse
Affiliation(s)
- Natalia D Magnani
- Pulmonary and Critical Care Division, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Laura A Dada
- Pulmonary and Critical Care Division, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Jacob I Sznajder
- Pulmonary and Critical Care Division, Northwestern Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
39
|
Sim TY, Harith HH, Tham CL, Md Hashim NF, Shaari K, Sulaiman MR, Israf DA. The Protective Effects of a Synthetic Geranyl Acetophenone in a Cellular Model of TNF-α-Induced Pulmonary Epithelial Barrier Dysfunction. Molecules 2018; 23:molecules23061355. [PMID: 29874809 PMCID: PMC6100020 DOI: 10.3390/molecules23061355] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/19/2018] [Accepted: 05/25/2018] [Indexed: 01/19/2023] Open
Abstract
Alveolar epithelial barrier dysfunction contributes to lung edema and can lead to acute lung injury (ALI). The features include increased epithelial permeability, upregulation of inflammatory mediators and downregulation of junctional complex molecules; these changes are often induced by inflammation. tHGA is an acetophenone analogue with therapeutic potential in asthma. Its therapeutic potential in ALI is presently unknown. Herein, the effects of tHGA on epithelial barrier dysfunction were determined in TNF-α-induced human alveolar epithelial cells. The anti-inflammatory properties of tHGA were assessed by monocyte adhesion assay and analysis of MCP-1 and ICAM-1 expression. The epithelial barrier function was assessed by paracellular permeability and transepithelial electrical resistance (TEER) assays, and analysis of junctional complex molecules expression. To elucidate the mechanism of action, the effects of tHGA on the NF-κB and MAPK pathways were determined. Gene and protein expression were analyzed by RT-PCR and Western blotting or ELISA, respectively. tHGA suppressed leukocyte adhesion to TNF-α-induced epithelium and reduced MCP-1 and ICAM-1 gene expression and secretion. tHGA also increased TEER readings, reduced epithelial permeability and enhanced expression of junctional complex molecules (zona occludens-1, occludin and E-cadherin) in TNF-α-induced cells. Correspondingly, the NF-κB, ERK and p38 MAPK pathways were also inhibited by tHGA. These findings suggest that tHGA is able to preserve alveolar epithelial barrier function in response to acute inflammation, via its anti-inflammatory activity and stabilization of epithelial barrier integrity, mediated by NF-κB, ERK and p38 MAPK signaling.
Collapse
Affiliation(s)
- Tee Yee Sim
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Hanis Hazeera Harith
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Nur Fariesha Md Hashim
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Khozirah Shaari
- Natural Products Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Mohd Roslan Sulaiman
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| |
Collapse
|
40
|
Yang CY, Chen CS, Yiang GT, Cheng YL, Yong SB, Wu MY, Li CJ. New Insights into the Immune Molecular Regulation of the Pathogenesis of Acute Respiratory Distress Syndrome. Int J Mol Sci 2018; 19:ijms19020588. [PMID: 29462936 PMCID: PMC5855810 DOI: 10.3390/ijms19020588] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/11/2018] [Accepted: 02/14/2018] [Indexed: 12/11/2022] Open
Abstract
Acute respiratory distress syndrome is an inflammatory disease characterized by dysfunction of pulmonary epithelial and capillary endothelial cells, infiltration of alveolar macrophages and neutrophils, cell apoptosis, necroptosis, NETosis, and fibrosis. Inflammatory responses have key effects on every phase of acute respiratory distress syndrome. The severe inflammatory cascades impaired the regulation of vascular endothelial barrier and vascular permeability. Therefore, understanding the relationship between the molecular regulation of immune cells and the pulmonary microenvironment is critical for disease management. This article reviews the current clinical and basic research on the pathogenesis of acute respiratory distress syndrome, including information on the microenvironment, vascular endothelial barrier and immune mechanisms, to offer a strong foundation for developing therapeutic interventions.
Collapse
Affiliation(s)
- Chin-Yao Yang
- Division of Chest Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Chien-Sheng Chen
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Yeung-Leung Cheng
- Division of Thoracic Surgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- School of Surgery, Tzu Chi University, Hualien 970, Taiwan.
| | - Su-Boon Yong
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.
- Division of Pediatric Allergy, Immunology and Rheumatology, Department of Pediatrics, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- Department of Nursing, Meiho University, Pingtung 912, Taiwan.
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| |
Collapse
|
41
|
Vyas-Read S, Vance RJ, Wang W, Colvocoresses-Dodds J, Brown LA, Koval M. Hyperoxia induces paracellular leak and alters claudin expression by neonatal alveolar epithelial cells. Pediatr Pulmonol 2018; 53:17-27. [PMID: 29168340 PMCID: PMC5938176 DOI: 10.1002/ppul.23681] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 01/03/2017] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Premature neonates frequently require oxygen supplementation as a therapeutic intervention that, while necessary, also exposes the lung to significant oxidant stress. We hypothesized that hyperoxia has a deleterious effect on alveolar epithelial barrier function rendering the neonatal lung susceptible to injury and/or bronchopulmonary dysplasia (BPD). MATERIALS AND METHODS We examined the effects of exposure to 85% oxygen on neonatal rat alveolar barrier function in vitro and in vivo. Whole lung was measured using wet-to-dry weight ratios and bronchoalveolar lavage protein content and cultured primary neonatal alveolar epithelial cells (AECs) were measured using transepithelial electrical resistance (TEER) and paracellular flux measurements. Expression of claudin-family tight junction proteins, E-cadherin and the Snail transcription factor SNAI1 were measured by Q-PCR, immunoblot and confocal immunofluorescence microscopy. RESULTS Cultured neonatal AECs exposed to 85% oxygen showed impaired barrier function. This oxygen-induced increase in paracellular leak was associated with altered claudin expression, where claudin-3 and -18 were downregulated at both the mRNA and protein level. Claudin-4 and -5 mRNA were also decreased, although protein expression of these claudins was largely maintained. Lung alveolarization and barrier function in vivo were impaired in response to hyperoxia. Oxygen exposure also significantly decreased E-cadherin expression and induced expression of the SNAI1 transcription factor in vivo and in vitro. CONCLUSIONS These data support a model in which hyperoxia has a direct impact on alveolar tight and adherens junctions to impair barrier function. Strategies to antagonize the effects of high oxygen on alveolar junctions may potentially reverse this deleterious effect.
Collapse
Affiliation(s)
- Shilpa Vyas-Read
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Rachel J Vance
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Wenyi Wang
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | | | - Lou Ann Brown
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Michael Koval
- Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, Georgia.,Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
42
|
Cai J, Culley MK, Zhao Y, Zhao J. The role of ubiquitination and deubiquitination in the regulation of cell junctions. Protein Cell 2017; 9:754-769. [PMID: 29080116 PMCID: PMC6107491 DOI: 10.1007/s13238-017-0486-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022] Open
Abstract
Maintenance of cell junctions plays a crucial role in the regulation of cellular functions including cell proliferation, permeability, and cell death. Disruption of cell junctions is implicated in a variety of human disorders, such as inflammatory diseases and cancers. Understanding molecular regulation of cell junctions is important for development of therapeutic strategies for intervention of human diseases. Ubiquitination is an important type of post-translational modification that primarily regulates endogenous protein stability, receptor internalization, enzyme activity, and protein-protein interactions. Ubiquitination is tightly regulated by ubiquitin E3 ligases and can be reversed by deubiquitinating enzymes. Recent studies have been focusing on investigating the effect of protein stability in the regulation of cell-cell junctions. Ubiquitination and degradation of cadherins, claudins, and their interacting proteins are implicated in epithelial and endothelial barrier disruption. Recent studies have revealed that ubiquitination is involved in regulation of Rho GTPases’ biological activities. Taken together these studies, ubiquitination plays a critical role in modulating cell junctions and motility. In this review, we will discuss the effects of ubiquitination and deubiquitination on protein stability and expression of key proteins in the cell-cell junctions, including junction proteins, their interacting proteins, and small Rho GTPases. We provide an overview of protein stability in modulation of epithelial and endothelial barrier integrity and introduce potential future search directions to better understand the effects of ubiquitination on human disorders caused by dysfunction of cell junctions.
Collapse
Affiliation(s)
- Junting Cai
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Miranda K Culley
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yutong Zhao
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jing Zhao
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Asthma, and Critical Care Medicine, Department of Medicine, The University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
43
|
Wynne BM, Zou L, Linck V, Hoover RS, Ma HP, Eaton DC. Regulation of Lung Epithelial Sodium Channels by Cytokines and Chemokines. Front Immunol 2017; 8:766. [PMID: 28791006 PMCID: PMC5524836 DOI: 10.3389/fimmu.2017.00766] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 06/16/2017] [Indexed: 12/20/2022] Open
Abstract
Acute lung injury leading to acute respiratory distress (ARDS) is a global health concern. ARDS patients have significant pulmonary inflammation leading to flooding of the pulmonary alveoli. This prevents normal gas exchange with consequent hypoxemia and causes mortality. A thin fluid layer in the alveoli is normal. The maintenance of this thin layer results from fluid movement out of the pulmonary capillaries into the alveolar interstitium driven by vascular hydrostatic pressure and then through alveolar tight junctions. This is then balanced by fluid reabsorption from the alveolar space mediated by transepithelial salt and water transport through alveolar cells. Reabsorption is a two-step process: first, sodium enters via sodium-permeable channels in the apical membranes of alveolar type 1 and 2 cells followed by active extrusion of sodium into the interstitium by the basolateral Na+, K+-ATPase. Anions follow the cationic charge gradient and water follows the salt-induced osmotic gradient. The proximate cause of alveolar flooding is the result of a failure to reabsorb sufficient salt and water or a failure of the tight junctions to prevent excessive movement of fluid from the interstitium to alveolar lumen. Cytokine- and chemokine-induced inflammation can have a particularly profound effect on lung sodium transport since they can alter both ion channel and barrier function. Cytokines and chemokines affect alveolar amiloride-sensitive epithelial sodium channels (ENaCs), which play a crucial role in sodium transport and fluid reabsorption in the lung. This review discusses the regulation of ENaC via local and systemic cytokines during inflammatory disease and the effect on lung fluid balance.
Collapse
Affiliation(s)
- Brandi M Wynne
- Department of Medicine, Nephrology, Emory University, Atlanta, GA, United States.,Department of Physiology, Emory University, Atlanta, GA, United States.,The Center for Cell and Molecular Signaling, Emory University, Atlanta, GA, United States
| | - Li Zou
- Department of Physiology, Emory University, Atlanta, GA, United States
| | - Valerie Linck
- Department of Physiology, Emory University, Atlanta, GA, United States
| | - Robert S Hoover
- Department of Medicine, Nephrology, Emory University, Atlanta, GA, United States.,Department of Physiology, Emory University, Atlanta, GA, United States.,Research Service, Atlanta Veteran's Administration Medical Center, Decatur, GA, United States
| | - He-Ping Ma
- Department of Physiology, Emory University, Atlanta, GA, United States.,The Center for Cell and Molecular Signaling, Emory University, Atlanta, GA, United States
| | - Douglas C Eaton
- Department of Physiology, Emory University, Atlanta, GA, United States.,The Center for Cell and Molecular Signaling, Emory University, Atlanta, GA, United States
| |
Collapse
|
44
|
Suliman HB, Kraft B, Bartz R, Chen L, Welty-Wolf KE, Piantadosi CA. Mitochondrial quality control in alveolar epithelial cells damaged by S. aureus pneumonia in mice. Am J Physiol Lung Cell Mol Physiol 2017; 313:L699-L709. [PMID: 28663335 DOI: 10.1152/ajplung.00197.2017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 12/30/2022] Open
Abstract
Mitochondrial damage is often overlooked in acute lung injury (ALI), yet most of the lung's physiological processes, such as airway tone, mucociliary clearance, ventilation-perfusion (Va/Q) matching, and immune surveillance require aerobic energy provision. Because the cell's mitochondrial quality control (QC) process regulates the elimination and replacement of damaged mitochondria to maintain cell survival, we serially evaluated mitochondrial biogenesis and mitophagy in the alveolar regions of mice in a validated Staphylococcus aureus pneumonia model. We report that apart from cell lysis by direct contact with microbes, modest epithelial cell death was detected despite significant mitochondrial damage. Cell death by TdT-mediated dUTP nick-end labeling staining occurred on days 1 and 2 postinoculation: apoptosis shown by caspase-3 cleavage was present on days 1 and 2, while necroptosis shown by increased levels of phospho- mixed lineage kinase domain-like protein (MLKL) and receptor-interacting serine/threonine-protein kinase 1 (RIPK1) was present on day 1 Cell death in alveolar type I (AT1) cells assessed by bronchoalveolar lavage fluid receptor for advanced glycation end points (RAGE) levels was high, yet AT2 cell death was limited while both mitochondrial biogenesis and mitophagy were induced. These mitochondrial QC mechanisms were evaluated mainly in AT2 cells by localizing increases in citrate synthase content, increases in nuclear mitochondrial biogenesis regulators nuclear respiratory factor-1 (NRF-1) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), and increases in light chain 3B protein (LC3-I)/LC3II ratios. Concomitant changes in p62, Pink 1, and Parkin protein levels indicated activation of mitophagy. By confocal microscopy, mitochondrial biogenesis and mitophagy were often observed on day 1 within the same AT2 cells. These findings imply that mitochondrial QC activation in pneumonia-damaged AT2 cells promotes cell survival in support of alveolar function.
Collapse
Affiliation(s)
- Hagir B Suliman
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Bryan Kraft
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Raquel Bartz
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Lingye Chen
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Karen E Welty-Wolf
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Claude A Piantadosi
- Departments of Medicine, Pathology, and Anesthesiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
45
|
Lee B, Kang HY, Lee DO, Ahn C, Jeung EB. Claudin-1, -2, -4, and -5: comparison of expression levels and distribution in equine tissues. J Vet Sci 2017; 17:445-451. [PMID: 27030194 PMCID: PMC5204021 DOI: 10.4142/jvs.2016.17.4.445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 02/27/2016] [Accepted: 03/04/2016] [Indexed: 11/24/2022] Open
Abstract
Claudins, which are known as transmembrane proteins play an essential role in tight junctions (TJs) to form physical barriers and regulate paracellular transportation. To understand equine diseases, it is helpful to measure the tissue-specific expression of TJs in horses. Major equine diseases such as colic and West Nile cause damage to TJs. In this study, the expression level and distribution of claudin-1, -2, -4, and -5 in eight tissues were assessed by Western blotting and immunohistochemistry methods. Claudin-1 was primarily identified in the lung, duodenum, and uterus, claudin-2 was evenly observed in equine tissues, claudin-4 was abundantly detected in the liver, kidney and uterus, and claudin-5 was strongly expressed in the lung, duodenum, ovary, and uterus, as determined by Western blotting method. The localization of equine claudins was observed by immunohistochemistry methods. These findings provide knowledge regarding the expression patterns and localization of equine claudins, as well as valuable information to understand tight junction-related diseases according to tissue specificity and function of claudins in horses.
Collapse
Affiliation(s)
- Bonn Lee
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Hee Young Kang
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Dong Oh Lee
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Changhwan Ahn
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
46
|
Peter A, Fatykhova D, Kershaw O, Gruber AD, Rueckert J, Neudecker J, Toennies M, Bauer TT, Schneider P, Schimek M, Eggeling S, Suttorp N, Hocke AC, Hippenstiel S. Localization and pneumococcal alteration of junction proteins in the human alveolar-capillary compartment. Histochem Cell Biol 2017; 147:707-719. [PMID: 28247028 DOI: 10.1007/s00418-017-1551-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2017] [Indexed: 02/03/2023]
Abstract
Loss of alveolar barrier function with subsequent respiratory failure is a hallmark of severe pneumonia. Although junctions between endo- and epithelial cells regulate paracellular fluid flux, little is known about their composition and regulation in the human alveolar compartment. High autofluorescence of human lung tissue in particular complicates the determination of subcellular protein localization. By comparing conventional channel mode confocal imaging with spectral imaging and linear unmixing, we demonstrate that background fluorescent spectra and fluorophore signals could be rigorously separated resulting in complete recovery of the specific signal at a high signal-to-noise ratio. Using this technique and Western blotting, we show the expression patterns of tight junction proteins occludin, ZO-1 as well as claudin-3, -4, -5 and -18 and adherence junction protein VE-cadherin in naive or Streptococcus pneumoniae-infected human lung tissue. In uninfected tissues, occludin and ZO-1 formed band-like structures in alveolar epithelial cells type I (AEC I), alveolar epithelial cells type II (AEC II) and lung capillaries, whereas claudin-3, -4 and -18 were visualised in AEC II. Claudin-5 was detected in the endothelium only. Claudin-3, -5, -18 displayed continuous band-like structures, while claudin-4 showed a dot-like expression. Pneumococcal infection reduced alveolar occludin, ZO-1, claudin-5 and VE-cadherin but did not change the presence of claudin-3, -4 and -18. Spectral confocal microscopy allows for the subcellular structural analysis of proteins in highly autofluorescent human lung tissue. The thereby observed deterioration of lung alveolar junctional organisation gives a structural explanation for alveolar barrier disruption in severe pneumococcal pneumonia.
Collapse
Affiliation(s)
- Andrea Peter
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Department for Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355, Berlin, Germany
| | - Diana Fatykhova
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Jens Rueckert
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Jens Neudecker
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Mario Toennies
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Torsten T Bauer
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Paul Schneider
- Department for General and Thoracic Surgery, DRK Clinics, Drontheimer Strasse 39-40, 13359, Berlin, Germany
| | - Maria Schimek
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Stephan Eggeling
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas C Hocke
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
47
|
Wittekindt OH. Tight junctions in pulmonary epithelia during lung inflammation. Pflugers Arch 2017; 469:135-147. [PMID: 27921210 PMCID: PMC5203840 DOI: 10.1007/s00424-016-1917-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/31/2022]
Abstract
Inflammatory lung diseases like asthma bronchiale, chronic obstructive pulmonary disease and allergic airway inflammation are widespread public diseases that constitute an enormous burden to the health systems. Mainly classified as inflammatory diseases, the treatment focuses on strategies interfering with local inflammatory responses by the immune system. Inflammatory lung diseases predispose patients to severe lung failures like alveolar oedema, respiratory distress syndrome and acute lung injury. These life-threatening syndromes are caused by increased permeability of the alveolar and airway epithelium and exudate formation. However, the mechanism underlying epithelium barrier breakdown in the lung during inflammation is elusive. This review emphasises the role of the tight junction of the airway epithelium as the predominating structure conferring epithelial tightness and preventing exudate formation and the impact of inflammatory perturbations on their function.
Collapse
Affiliation(s)
- Oliver H Wittekindt
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
48
|
Barmeyer C, Fromm M, Schulzke JD. Active and passive involvement of claudins in the pathophysiology of intestinal inflammatory diseases. Pflugers Arch 2016; 469:15-26. [PMID: 27904960 DOI: 10.1007/s00424-016-1914-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/20/2016] [Accepted: 11/22/2016] [Indexed: 12/18/2022]
Abstract
Intestinal inflammatory diseases, four of which are discussed here, are associated with alterations of claudins. In ulcerative colitis, diarrhea and antigen entry into the mucosa occurs. Claudin-2 is upregulated but data on other claudins are still limited or vary (e.g., claudin-1 and -4). Apart from that, tight junction changes contribute to diarrhea via a leak flux mechanism, while protection against antigen entry disappears behind epithelial gross lesions (erosions) and apoptotic foci. Crohn's disease is additionally characterized by a claudin-5 and claudin-8 reduction which plays an active role in antigen uptake already before gross lesions appear. In microscopic colitis (MC), upregulation of claudin-2 expression is weak and a reduction in claudin-4 may be only passively involved, while sodium malabsorption represents the main diarrheal mechanism. However, claudin-5 is removed from MC tight junctions which may be an active trigger for inflammation through antigen uptake along the so-called leaky gut concept. In celiac disease, primary barrier defects are discussed in the context of candidate genes as PARD3 which regulate cell polarity and tight junctions. The loss of claudin-5 allows small antigens to invade, while the reductions in others like claudin-3 are rather passive events. Taken together, the specific role of single tight junction proteins for the onset and perpetuation of inflammation and the recovery from these diseases is far from being fully understood and is clearly dependent on the stage of the disease, the background of the other tight junction components, the transport activity of the mucosa, and the presence of other barrier features like gross lesions, an orchestral interplay which is discussed in this article.
Collapse
Affiliation(s)
- Christian Barmeyer
- Institute of Clinical Physiology, Charité-Universitätsmedizin Berlin, 12203, Berlin, Germany
| | - Michael Fromm
- Institute of Clinical Physiology, Charité-Universitätsmedizin Berlin, 12203, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology, Charité-Universitätsmedizin Berlin, 12203, Berlin, Germany.
| |
Collapse
|
49
|
Zheng Y, Cai W, Zhou S, Xu L, Jiang C. Protective effect of bone marrow derived mesenchymal stem cells in lipopolysaccharide-induced acute lung injury mediated by claudin-4 in a rat model. Am J Transl Res 2016; 8:3769-3779. [PMID: 27725857 PMCID: PMC5040675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/04/2016] [Indexed: 06/06/2023]
Abstract
Our study aims to investigate the effects of bone marrow derived mesenchymal stem cells (BM-MSCs) in lipopolysaccharide (LPS)-induced acute lung injury (ALI) as well as the underlying mechanism. In our study, Wistar rats were randomly divided into four groups: control group; ALI group; ALI+MSCs group and ALI+MSCs claudin-4 siRNA group. MRC-5 and BEAS-2B cell lines were used for in vitro assay. Flow cytometry, western blot, hematoxylin and eosin (H&E) staining, CCK-8 assay, enzyme-linked immunosorbent assay (ELISA) were involved to measure the pathological changes in lung tissues. Results showed that in vivo MSCs administration significantly attenuated pulmonary edema (wet/dry ratio), inflammation cytokines levels (TGF-α), pathological alternations and cell apoptosis which were mediated by claudin-4 in LPS-induced acute lung injury in rats. In vitro experiment showed that hypoxia could induce the expression of claudin-4 in MSCs, and MSCs treatment showed significantly enhanced cell viability (by CCK-8 assay) and reduced cell apoptosis. In conclusion, the present study demonstrated that BM-MSCs can protect against LPS-induced ALI in vivo and in vitro, at least partly mediated by claudin-4.
Collapse
Affiliation(s)
- Yueliang Zheng
- Department of Emergency, Zhejiang Provincial People's Hospital Hangzhou 310014, China
| | - Wenwei Cai
- Department of Emergency, Zhejiang Provincial People's Hospital Hangzhou 310014, China
| | - Shengang Zhou
- Department of Emergency, Zhejiang Provincial People's Hospital Hangzhou 310014, China
| | - Liming Xu
- Department of Emergency, Zhejiang Provincial People's Hospital Hangzhou 310014, China
| | - Chengxing Jiang
- Department of Emergency, Zhejiang Provincial People's Hospital Hangzhou 310014, China
| |
Collapse
|
50
|
Kielgast F, Schmidt H, Braubach P, Winkelmann VE, Thompson KE, Frick M, Dietl P, Wittekindt OH. Glucocorticoids Regulate Tight Junction Permeability of Lung Epithelia by Modulating Claudin 8. Am J Respir Cell Mol Biol 2016; 54:707-17. [DOI: 10.1165/rcmb.2015-0071oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|